Biobank want to make it hard to access their project information — so it's all in this file

In July 2024 UK Biobank stopped publishing details of new projects. They kept approving customers who wanted to start new projects, they just kept the details secret until July 2025 when Biobank disclosed projects on their "new website", a new website which had the intermittent and unresolved effect of blocks services like the Internet Archive from taking an independent copy of what UK Biobank said then. (UKBiobank have told us this is the IA's fault, they they believe they have unblocked the IA from those they were intending to block from accessing their website. UKB now allows a small number of links to be archived, and then intermittently blocks them). Biobank keeps secret which projects have been given exceptions to download the data and use it without any oversight. Biobank also keep secret what data a project has been granted access to.

While Biobank were in their period of coverup, their former "Chief Scientist" talked about Biobank in glowing terms at the launch of the HDR/Sudlow Review on the future of research data, proclaiming Biobank gives data out in "days" and proclaimed Biobank have "one of the best systems" for giving data out rapidly. Biobank's recent annual report discloses they give exceptions to their data governance rules — have they given any exceptions to their "Pioneer Fund" affiliated users? How about to the company which runs tiktok? (Biobank say it's a US company, which seems... fishy?)

That HDR/Review proposed the Health Data Research Service, which presumably will send data to the eugenicists, the sanctioned entities, and the shell companies that remain on the UK Biobank project list.

Not that you'd easily be able to tell.

UK Biobank continues to actively configure their systems to block most independent archiving of their website – UKB claim it's not their fault, and they're no more competent at technology than they are at checking the short forms they have decided to use to grant access to half a million people's DNA sequences and health records...

UK Biobank also refuses to say what data/material individual projects requested and were given, or whether individual projects have been given TRE exceptions. In theory UKB's public statements mean most projects could be excepted from the rules they claim to have.

The best way to understand how a Service will act tomorrow is to understand how it acts today. The HDR Service conceived by the HDR/Sudlow Review is likely to perpetuate a culture of coverup and risk taking with data, simply because that's what UK Biobank has decided is acceptable.

After months of conversations, Biobank still hides from scrutiny, so we regularly build and publish a 2500+ page PDF and an equivalent spreadsheet of all their projects until they don't wish to hide any more. This is the contents of that file.

medConfidential

 logo

This file was built at Mon Nov 10 10:22:10 2025.










Projects which seem to have disappeared:





Newly listed projects this month


Some of these may be old projects being newly disclosed or redisclosed.



https://www.ukbiobank.ac.uk/projects/10-year-cardiovascular-risk-calculator-using-conventional-genetic-and-social-data

10-year cardiovascular risk calculator using conventional, genetic, and social data

Last updated:
ID:
179482
Start date:
28 March 2024
Project status:
Current
Principal investigator:
Dr Nikita Umov
Lead institution:
University of Tartu, Estonia

Heart attacks and strokes are big health problems around the world. They are the main reason why people die. Whether someone might get these diseases depends on different things. There are some factors that people can change. These include how much cholesterol is in your blood, blood pressure, how much you exercise, weight, educational qualifications, whether you smoke, whether you are rich or poor, and how much time you spend with friends or family.
There are also factors that people can’t change. This includes age, whether you are male or female, ethnic background, genes, and whether your family has a history of heart disease.
We will create a heart disease risk calculator. This uses information about cholesterol and blood pressure, genetic information and social factors. This tool will help predict how likely someone will get heart disease in the next ten years.
By using information about a person’s genes and social background, we can make this tool more accurate than the ones we have now. Getting an accurate prediction of your risk of heart disease is vital. Knowing the risk of getting heart disease allows you and your healthcare provider to make better choices about living healthier, like changing your diet or starting new medicines.
This research could change the way we treat heart diseases. It might lead to more personal and effective ways to prevent these diseases, helping reduce how often they happen and making people healthier.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/1000-ancient-british-genomes-and-the-uk-biobank-evolution-history-and-disease

1,000 ancient British genomes and the UK BioBank: evolution, history, and disease

Last updated:
ID:
89109
Start date:
3 November 2022
Project status:
Current
Principal investigator:
Dr Pontus Skoglund
Lead institution:
The Francis Crick Institute Limited, Great Britain

The British population is currently the world’s most well-studied in the field of human medical genetics. This 5-year project extends the UK Biobank to a 3rd dimension, by adding DNA from more than 1,000 individuals who lived in Britain during the past 4,500 years. Performing the first fine-scale studies of ancient DNA from a single place will allow us to understand human evolution in response to epidemics, dietary shifts, urbanisation, and industrialisation. It will also aid archaeology and medical genetics by untangling the complex and rich ancestry of present-day British populations from Stone Age, through the Middle Ages to today. Collaborating closely with archaeologists, the project will contextualise DNA information about studied individuals, creating new ways to understand the complex tapestry of ancestry through the human past. By understanding how human biology evolved in the past, we will be better prepared to tackle health challenges today.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/3d-reconstruction-of-human-anatomy-structure-from-medical-imagining-and-text-description

3D reconstruction of Human Anatomy Structure from Medical Imagining and Text Description

Last updated:
ID:
103180
Start date:
30 May 2023
Project status:
Current
Principal investigator:
Professor Xiaosong Yang
Lead institution:
Bournemouth University, Great Britain

Traditional medical imaging, such as X-rays, CT scans, and MRI scans, have been instrumental in diagnosing and treating a wide variety of medical conditions. However those 2D image or 3D volume data are difficult to interpret and visualize in a way that accurately represents the full 3D structure of the body. This can make it challenging for medical professionals to get a complete picture of a patient’s anatomy and identify abnormalities. This inspired lots of research in the past 20 years on medical image segmentation and 3D visualization. Given high resolution imagining and dense scan, the state-of-the-art techniques can reconstruct the 3D model accurately.
However, many medical imaging methods, such as X-rays and CT scans, use ionizing radiation to create images. While the doses used are generally considered safe, repeated exposure can increase the risk of cancer or other radiation-related health problems. Some medical imaging methods can be time-consuming and require patients to lie still for an extended period of time. This can be difficult for some patients, such as those with claustrophobia or mobility issues. Besides, medical imaging methods can be expensive in terms of the equipment and staff cost. This leads to only a very limited imaging could be acquired for each patient, which is normally not enough for the 3D reconstruction to show a complete visualization of the patient’s problem.

Our goal is to reduce the number of medical image captures and supply a 3D visualization of patient’s anatomy using machine learning techniques. Medical data are normally multimodality (image, text, audio, video etc), complex with information from various channels. Machine learning technique have been proven very efficient in processing large data, learn their structures, extract useful features, perform automated analysis, enabling faster and more consistent reasoning. This can improve efficiency in medical workflows, reduce errors, and facilitate more informed decision-making by healthcare providers.
Our project aims to develop a few new machine learning models to 1) train a parametric 3D human skeleton model that can represent individual patient skeletal structure, and 2) efficiently and accurately reconstruct 3D human anatomy from medical images and report.
Our project is expected to last three years and will have a significant impact on productivity in the NHS at scale. By enabling doctors to make more informed decisions and helping professionals and patients understand medical conditions and treatments, our technology has the potential to improve healthcare outcomes.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/3d-shape-generation-and-digital-reconstruction-of-head-and-trunk-organs-and-tissues

3D Shape Generation and Digital Reconstruction of Head and Trunk Organs and Tissues

Last updated:
ID:
482388
Start date:
26 June 2025
Project status:
Current
Principal investigator:
Dr Nan Zhang
Lead institution:
Chinese Academy of Sciences, China

1) 3D Reconstruction of Head and Trunk Skeletons and Soft Tissue Organs: Using full-body MRI and DEXA data from the UK Biobank, we plan to reconstruct the 3D shapes of the head and trunk skeletons, as well as major soft tissue organs (such as the lungs, liver, heart, spleen, artery, etc.).

2) Segmentation and Reconstruction of Brain Regions and Blood Vessels: Develop AI-based tools to automatically segment and reconstruct brain regions and blood vessels by analyzing structural MRI, MR angiography (MRA), ultrasound imaging (US), and computed tomography angiography (CTA) data. This will aid in identifying key brain areas, optimizing neurosurgical planning, and providing precise navigation support for cerebrovascular surgeries.

3) Generation of Internal Organ and Tissue Shapes Based on RGB Images: By analyzing the correlation between the shape of the skin surface and internal organs and tissues, we will develop a system for generating 3D shapes of internal organs and tissues based on RGB images. This system will utilize deep learning and advanced image processing techniques to infer the shapes of internal organs and tissues from external images.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-biologically-meaningful-and-interpretable-neural-network-framework-for-end-to-end-multi-phenotype-prediction

A biologically meaningful and interpretable neural network framework for end-to-end multi phenotype prediction

Last updated:
ID:
97487
Start date:
13 April 2023
Project status:
Current
Principal investigator:
Professor Yves Moreau
Lead institution:
Katholieke Universiteit Leuven, Belgium

This project aims to develop new tools to better predict a patient’s risk of genetic diseases. The tool is developed using machine learning techniques and makes a disease risk assessment based on the patient’s genetic data and existing biological knowledge. Besides providing a risk analysis, the project also aims to understand these risk predictions. By interpreting how and based on what genetic factors the prediction for patients is made, we aim to gain insides in the underlying disease mechanisms and which genetic factors are involved.
The project is part of a larger genome interpretation project carried out by the lab of Yves Moreau. The past decades faster and cheaper techniques to read out DNA have created a large amount of genetic data. This opens the possibility to investigate the complex relations between genetics and diseases by using sophisticated and data-hungry machine learning techniques. Ultimately, this will provide diagnostic tools that help clinicians to assess disease risk more precisely and lead to a deeper understanding of how genetic factors are causing diseases, possibly providing new treatment options.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-biopsychosocial-model-of-the-effect-of-social-support-on-mental-health-in-middle-to-older-aged-adults

A biopsychosocial model of the effect of social support on mental health in middle- to older-aged adults

Last updated:
ID:
54646
Start date:
14 May 2020
Project status:
Closed
Principal investigator:
Miss Jessica Stepanous
Lead institution:
University of Manchester, Great Britain

Social relationships give support and meaning in our lives, contributing to good mental health. However, the precise way in which social support leads to good mental health is unclear. There are several interacting factors to consider, such as the type of social support (Hakulinen et al., 2016), socioeconomic conditions (Berkman & Krishna, 2014), and an individual’s personality (Kendler et al., 2002). There is also a dynamic interplay between biological and social factors, as the structure and function of the brain affects receptivity to social support, and social support shapes neural structure and function (Lamblin et al., 2017). Previous research has looked at these different factors individually, however this project aims to build on this to combine biological, psychological and social factors into a model of the mental health benefits of social support.

A statistical method called structural equation modelling will be used to test different relationships between different types of social support on mental health outcomes, the impact of social and personality factors, and how it is related to the structure and function of the brain. The project will use data from the UK Biobank which includes extensive questionnaire data as well as brain imaging data in a very large sample of middle- to older-aged adults. It will form the first study of a PhD and last around 18 months.

The findings of the project could help to create new approaches to improving mental health. By understanding how the brain is affected by social factors and how this relates to anxiety and depression, we can develop new evidence-based approaches to treating and preventing mental health problems that focus on enhanced social support. Loneliness and social isolation is a rising concern, especially in older people, and it is essential to understand this at a biological as well as a social level.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-catalogue-of-missing-environmental-influences

A catalogue of ‘missing environmental influences’

Last updated:
ID:
58285
Start date:
8 January 2021
Project status:
Closed
Principal investigator:
Professor Felix Christian Tropf
Lead institution:
Center for Research in Economics and Statistics, France

All studied human traits are partly genetic and partly environmentally influenced. While
geneticists have gone to great lengths to find genes that predict human traits, social science has yet to catch up and consider the involvement of genes towards explaining aspects of the human condition. How well do we explain and predict health, education, fertility or mortality based on knowledge about someone’s behaviour (social) environment? How well do we explain and predict human behaviour based on our theories? How much of the social relationships we observe may actually be influenced by genetic causes?

In order to evaluate currently ongoing, costly large-scale data collections and research initiatives, we need a robust assessment to evaluate our reasoning about the social world, the quality of our policy recommendations, and timeless debates about nature versus nurture. The UKBiobank provides us with the unique opportunity to analyse amazingly rich information, whilst considering the interactions among genes and the environment. Central questions that we aim to answer are therefore: To what extent does the (social) environment explain and predict our behaviour or health? How well are we doing in collecting the right data? Do we need more advanced social theories, or do we need to do a better job at collecting data to improve our findings?

In this project, we will, over the course of 36 months, isolate environmental influences from genetic influences to quantify the importance of environmental factors that contribute to various health, status and demographic characteristics of individuals. Furthermore, we aim to evaluate whether future research requires more advanced thinking or more data when it comes to explaining or understanding human conditions. Results may question the stability of our current social science explanations, and highlight the need for complexity in our reasoning about the (social) world. Finally, we approach the complex interplay between genes and the environment and issues of genetic selection in order to approve substantial and statistical knowledge for general knowledge and future research directions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-clinical-event-prediction-model-linking-clinical-complications-of-obesity-with-associated-risk-factors

A Clinical Event Prediction Model Linking Clinical Complications of Obesity with Associated Risk Factors

Last updated:
ID:
128965
Start date:
6 December 2023
Project status:
Current
Principal investigator:
Mr Dennis Xuan
Lead institution:
Tulane University, United States of America

Aims: To produce a predictive outcome model framework with which to analyze and quantitatively characterize the associations of risk factors and outcomes in an obese population through multivariable regression analysis
Rationale: A risk prediction model of this type does not yet exist for the obese population which generally characterizes the links between risk factors and outcomes. Utilizing a large dataset available within the UK Biobank would serve as a foundation for the creation of such a tool and provide room for expansion and iterative improvement in the future.
Duration: 3 years with annual updates
Impact: Understanding and predicting the progression of obesity would be invaluable towards shaping adverse outcome prevention, care, and disease management. In addition, by basing the project in an ongoing data collection project, there is always the possibility of iterative improvement in the future.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-cohort-study-to-investigate-associations-between-herpes-zoster-or-herpes-simplex-related-disease-and-frailty-in-middle-age-and-older-adults

A cohort study to investigate associations between herpes zoster or herpes simplex-related disease and frailty in middle-age and older adults

Last updated:
ID:
320019
Start date:
29 November 2024
Project status:
Current
Principal investigator:
Mr Kousuke Iwai Saito
Lead institution:
Niigata University, Japan

Aim: To investigate whether herpes-zoster and herpes simplex virus-related diseases are associated with frailty
Scientific rationale: Recent reports showed that herpes zoster and herpes simplex viruses-related diseases were associated with neurodegenerative diseases including Alzheimer’s disease and cardiovascular diseases which are major risk factors of frailty in older adults. Herpes zoster and herpes simplex viruses -related disease are frequent in older adults because those people have decline of immunological function, which enable these viruses reactivate and cause related-diseases in those. However, it remains unknown whether those herpes virus-related diseases are associated with frailty.
Project duration: 2024 to 2029
Public health impact: Preventions of those herpes zoster and herpes simplex virus-related disease with vaccination or rapid prescription of medications against these herpes viruses will contribute to prevention of frailty in middle-age and older adults in case that the study show that those herpes virus-related diseases are associated with frailty in those.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-combined-risk-prediction-model-for-type-1-diabetes-mellitus

A combined risk prediction model for type 1 diabetes mellitus

Last updated:
ID:
77583
Start date:
9 August 2022
Project status:
Current
Principal investigator:
Professor Yilei Zhao
Lead institution:
Shanghai Jiao Tong University, China

Aim:
We aim to provide individualized risk prediction for type 1 diabetes mellitus (T1D) combining both clinical measurements and genetic data.

Scientific rationale:
T1D is a complex disease caused by both genetic and environmental factors such as lifestyles. Genetic factors include single-nucleotide changes that could alter functionality of proteins that could further lead to transition from physiological state to disease state. Many of the nucleotide changes have been discovered. A multitude of these changes have been found to be enriched in T1D patients compared to unaffected population.
Leveraging the rich datasets from UKBB, we aim to develop a prediction algorithm that would calculate individualized risk for T1D by combining effects from the nucleotide changes associated with T1D disease throughout the whole genome.
Additionally, we would investigate the interplay between genetics and clinical presentation including age, sex, glucose level, family history and biomarkers to further understand how genetics interacts with each component to influence disease susceptibility.

Project duration: 09/2021-09/2024
Public health impact:
The goal of developing a prediction model for T1D could potentially identify at-risk population for T1D when they are at pre- or early clinical stage. Our proposed model will add to the clinical tool box for risk prediction for T1D. It may later be applied to clinical consultation, disease management or personalized treatment. Implementation of this predictive scheme would potentially help clinicians to fine-tune the approach of caring or treating T1D patients to mitigate long-term impact on their lives.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-composite-approach-for-detecting-genomic-regions-under-local-adaptation-via-modeling-genotype-environment-interaction

A Composite approach for detecting genomic regions under local adaptation via modeling Genotype-Environment Interaction

Last updated:
ID:
96943
Start date:
22 July 2025
Project status:
Current
Principal investigator:
Professor Analabha Basu
Lead institution:
National Institute of Biomedical Genomics, India

Anatomically Modern Humans (AMH) originated and adapted in Africa for a long time before moving Out of Africa(OoA) around 50000-100000 years before present and underwent several demographic transitions in the journey of peopling the Globe. During this journey, AMH faced a different environment than the African subcontinent environment. Over time, AMH subpopulations moved into different parts of the globe and adapted to the specific local environment. This journey draws attention to the constant interaction of AMH populations with the environment i.e. Genotype-Environment(GxE) interaction. The constant GxE interactions resulted in spatially varying phenotypic traits across populations living in a local geographical space. This research work aims to investigate the role of GxE interaction in the local adaptation of human populations. The study aims to develop Machine Learning and Deep Learning model for detecting the genetic variants associated with environmental factors and selected for adaptive phenotypic traits. Identifying genetic and environmental factors responsible for the phenotypic trait variation across populations is crucial in public health and this study sheds light on some of the factors responsible for spatially varying phenotypic traits across human populations residing in a local geographical space.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-comprehensive-assessment-of-the-interactions-in-heart-brain-cross-talk-based-on-brain-structural-analyses

A comprehensive assessment of the interactions in heart-brain cross-talk based on brain structural analyses

Last updated:
ID:
653314
Start date:
12 May 2025
Project status:
Current
Principal investigator:
Dr Chengluan Xuan
Lead institution:
First Hospital of Jilin University, China

Research question: The magnitude of nervous system and brain structure changes induced by cardiovascular diseases is unclear.
Aims: We are committed to exploring unknown territory of heart-brain cross-talk, seeking out novel evidence and deciphering the underlying mechanisms. By doing so, we aim to establish new diagnostic approaches and formulate innovative therapeutic strategies that could potentially revolutionize the way we address the complex interplay between heart and brain health.
Scientific rationale: In cardiac dysfunction strong haemodynamics and neuronal signaling feedback interactions between heart and central nervous system exist that are able to bidirectionally provoke acute or chronic functional impairment. Cerebral injury secondary to cardiac dysfunction may include sudden stroke, cognitive decline (which might end up as dementia), and depression. Brain stem functions are related to the heart-brain interaction. In cardiac dysfunction, it’s known that the neurohormonal control and neuronal reflex circuits get out of balance. In this project, we will explore and identify the principal aspects of the pathophysiologic interactions between heart and brain that are associated with cardiac dysfunction. These aspects encompass a range of conditions and mechanisms, including stroke, the impact on cognitive function and brain structure, the relationship with depressive disorders, as well as alterations in neurovegetative control and the regulation of neuronal cardiovascular reflexes.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-comprehensive-benchmarking-analysis-of-fast-algorithms-for-genome-wide-association-studies

A comprehensive benchmarking analysis of fast algorithms for genome-wide association studies

Last updated:
ID:
281070
Start date:
27 November 2024
Project status:
Current
Principal investigator:
Dr Yong Jiang
Lead institution:
Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Germany

Genome-wide association study (GWAS) is a statistical tool for detecting associations between genetic variants and phenotypic traits. It is widely applied by scientists studying human, animal and plant genetics. During the past 15 years, many fast GWAS algorithms have been published to cope with the rapidly growing data size. But these algorithms do not necessarily yield consistent results for the same data set due to the implementation of distinct mathematical models and techniques. Thus, we can ask two questions: 1) What are the influence of the mathematical techniques implemented in different algorithms on the result of GWAS? 2) Given a certain set of parameters characterizing the data set (such as the population size, the complexity of genetic architecture), are some algorithms more suitable to apply than others? These questions are important because the results of GWAS suggest candidate regions in the genome which is usually a first step for finding causal genetic variant for important traits, such as complex deceases.
The aim of this project is to provide a knowledge-based solution to the two problems. First, we will provide an in-depth overview of key mathematical techniques implemented in commonly used fast GWAS algorithms. Then, we will select representative algorithms (Some algorithms may essentially implement the same technique despite that they are in different software packages) for the next step, namely a benchmarking analysis. We will compare the results of selected algorithms on empirical data sets across different species (among which human is of course an important one) and evaluate their computational efficiency (for this purpose data sets from UK Biobank is crucial because of its large scale). But since the causal genetic variant is largely unknown in empirical data sets, we will conduct a comprehensive simulation study to evaluate the statistical power and false positive rate of different algorithms. Based on real genomic data from different species, simulated traits with distinct population size, heritability and genetic architecture will be generated according to mathematical models, and each algorithm will be applied to each simulated data set. By combining the results of theoretical review, empirical data analyses and simulation study, we will be able to find out the answer to the two questions mentioned above. We expect to provide a summary on the pros and cons of different techniques implemented in the algorithms as well as recommendations on the choice of algorithms for the research community.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-comprehensive-evaluation-of-risk-factors-distinct-to-early-onset-colorectal-cancer

A comprehensive evaluation of risk factors distinct to early-onset colorectal cancer

Last updated:
ID:
154478
Start date:
7 March 2024
Project status:
Current
Principal investigator:
Dr Shria Kumar
Lead institution:
University of Miami, United States of America

Early onset colorectal cancer (EOCRC) is becoming more common, and it’s essential to understand what causes it. Several factors have been suggested as potential causes, including age, genetics, diet, physical activity, obesity, and more. Nuanced risk factors – like telomere length and body composition – have also been suggested, but are under-investigated. It’s important to comprehensively evaluate all these factors together, and also understand whether and how these risk factors impact EOCRC specifically (by comparing their impact on colorectal cancers in persons >50). This study aims to answer two important questions. First, it wants to understand the various factors that might lead to EOCRC, including telomere length and body composition. Second, we compare risk factors between EOCRC and colorectal cancer that occurs later in life. We will do this by conducting a large analysis of the UK Biobank data, using univariable and multivariable logistic regressions. We will explore a wide variety of risk factors, and evaluate their association with EOCRC and later-onset CRC. The project is expected to be completed and published within 24 months. The public health impact is substantial. EOCRCs have increased by >2% annually, and we need to understand risk factors so we can prevent and screen for EOCRC. We will use the generated results to then risk-stratify persons and ultimately, recommend screening strategies based on personalized risk.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-comprehensive-multiomic-examination-of-gastroenteropancreatic-neuroendocrine-neoplasms-gep-nens

A comprehensive multiomic examination of gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs).

Last updated:
ID:
194343
Start date:
3 October 2024
Project status:
Current
Principal investigator:
Dr Saiji Kathiresu Nageshwaran
Lead institution:
University College London, Great Britain

Gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) are a group of rare cancers that form in hormone-producing cells of the digestive system. The number of people diagnosed with these cancers is increasing worldwide, but we still don’t fully understand what causes them or how to best treat them.

Our research aims to substantially improve knowledge of the biological basis of GEP-NENs by analysing genetic, biochemical, imaging, lifestyle and health data from a large group of people. We will study information from an existing group of 800 GEP-NEN patients as well as from over 500,000 volunteers in the UK Biobank.

By comparing data from people with and without these cancers, we hope to validate known risk factors and discover new biological markers and treatment targets. We will use advanced statistical and computational methods to determine which factors are most important in causing GEP-NENs to develop and progress.

The scale and depth of data in the UK Biobank provides an unprecedented opportunity to make meaningful discoveries about these understudied cancers. Combining findings from our patient cohort with the Biobank data will allow us to develop robust predictive models to improve diagnosis and prognosis. We will also identify the most promising drug targets to accelerate development of new therapies.

Importantly, we will apply rigorous analysis methods to ensure the reliability of the risk factors and biological targets identified. All data and findings will be shared openly with the research community to maximise scientific impact.

This 3-year project will generate high-impact publications and data resources that substantially advance understanding of what drives GEP-NENs. The most significant public health impact will come from discovering new biomarkers for earlier diagnosis and treatment response monitoring, and from identifying validated drug targets to catalyse development of better therapies to improve and extend patients’ lives.

Leveraging the power of the UK Biobank in combination with our unique patient cohort will help transform outcomes for people with these rare but devastating cancers. The rigorous approach ensures the results will have maximal scientific impact and clinical relevance to drive much-needed progress against GEP-NENs.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-comprehensive-study-on-risk-and-protective-factors-of-sarcopenia-and-its-transition-through-clinical-and-multi-omics-approaches

A Comprehensive Study on Risk and Protective Factors of Sarcopenia and Its Transition Through Clinical and Multi-Omics Approaches

Last updated:
ID:
536492
Start date:
3 January 2025
Project status:
Current
Principal investigator:
Professor Miji Kim
Lead institution:
Kyung Hee University, Korea (South)

Sarcopenia, characterized by the age-related loss of muscle mass and function, is closely associated with increased risks of falls, hospitalizations, and mortality in older adults. Early detection of sarcopenia is crucial to mitigate its progression and minimize its impact on health outcomes.
The etiology of sarcopenia is both multifactorial and complex. Numerous factors, including malnutrition, physical inactivity, age-related cellular changes, oxidative stress, inflammation, and hormonal imbalances, contribute to the age-related decline in muscle mass and strength with age. This complexity, along with its association with various other health-related conditions, has driven growing interest in identifying reliable biomarkers to monitor its progression.
To date, most reported studies have primarily focused on identifying single biomarkers for sarcopenia. However, given the “multifactorial” nature of sarcopenia involving multiple biological pathways, relying on a single marker is unlikely to provide sufficient or reliable information. Instead, a multifaceted approach that integrates clinical data, biological insights, and multi-omics analysis is essential for accurately classifying and assessing older adults with sarcopenia.
Despite growing interest, a significant research gap persists, particularly in studies combining clinical data and multi-omics analysis through artificial intelligence (AI). Addressing this gap, our three-year project aims to uncover novel risk factors for sarcopenia by utilizing clinical data, multi-omics insights, and advanced AI methodologies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-comprehensive-study-on-the-causal-factors-early-diagnosis-and-screening-of-malignant-cancer

A comprehensive study on the causal factors, early diagnosis, and screening of malignant cancer.

Last updated:
ID:
226006
Start date:
3 October 2024
Project status:
Current
Principal investigator:
Professor Min Zhang
Lead institution:
Tsinghua University, China

Aims:
Cancer is a leading global cause of death. Nearly half of all cancers can be prevented through the avoidance of risk factors and the implementation of prevention strategies. Furthermore, early diagnosis and timely treatment increase the chances of curing many types of cancer. This research aims to identify the factors responsible for genetic changes that lead to cancer and to create early detection methods for various cancer types.

Scientific rationale:
Cancer evolves through stages, starting from normal cells transforming into pre-cancerous lesions and eventually forming malignant tumors. This multi-stage process is influenced by genetic and environmental factors interactions. Identifying the interplay of these genetic and environmental causal factors is crucial for effective cancer prevention. Moreover, reducing cancer mortality is more likely when cases are identified and treated early. Early detection is mainly based on two key approaches: early diagnosis and screening. Early diagnosis relies on identifying early symptoms of different forms of cancer. The early symptoms consist of changes in the characteristics of organs with lesions and adverse impacts on cognitive functions, physical ability, visual acuity, etc. On the other hand, the goal of screening is to pinpoint individuals with specific pre-cancer before symptoms appear. Screening is more resource-intensive than early diagnosis, demanding more precise targeted population identification and biomarker threshold detections. The identification of the cancer causal factors and the development of early cancer detection methods involve a thorough review of existing literature; integrating data from various sources like genomics, biomarkers, environment data, imaging data; using machine learning and statistical methods. The UK Biobank will be the source of the dataset.

Project duration:
We are proposing a project duration of 36 months for our research. The consideration of the timeframe is based on the time needed for processing and preparing the dataset for analysis, conducting statistical analyses, and drafting manuscripts for publication. We believe this duration will allow us to conduct a thorough and comprehensive investigation, ensuring the quality and impact of our research findings.

Public health impact:
The expected impact of the research is its potential to prevent cancers through targeted risk reduction and increase cure rates by enabling earlier intervention. By addressing crucial aspects of cancer control, the research aims to lessen the burden of cancer on individuals and healthcare systems.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-comprehensive-study-to-explore-neuroendocrine-biomarkers-differentially-expressed-in-patients-with-major-depressive-disorder-and-their-implications-for-disease-diagnosis-and-response-to-antidepressa

A comprehensive study to explore neuroendocrine biomarkers differentially expressed in patients with major depressive disorder and their implications for disease diagnosis and response to antidepressa

Last updated:
ID:
193969
Start date:
27 May 2025
Project status:
Current
Principal investigator:
Dr Ritushree Kukreti
Lead institution:
CSIR-Institute of Genomics and Integrative Biology, India

Early diagnosis of MDD contributes to a rapid and effective intervention and treatment process and thus is in high demand. Structured clinical interview is currently the most dominant way to diagnose MDD and track the treatment response, where the physician determines the patient’s symptoms by verbally interacting with the patient using standardized assessment tools, which are either clinician-administered or sometimes self-rated. Nevertheless, because the diagnostic process is relatively subjective, dependent on the physician’s expertise and the patient’s cooperation, subjected to human factors, and usually time-consuming, its applicability in the population and its diagnostic effectiveness are not ideal. Precise diagnosis of MDD through biochemical tests remains challenging due to the lack of objective physiological indicators, as well as specific laboratory tests. This indicates the need to develop more effective diagnosis methods based on an in-depth understanding of MDD’s pathophysiology. While no single biomarker exists for MDD diagnosis, there is mounting evidence of multiple dysregulated contributing factors, including inflammatory cytokines, endocrine factors, growth factors, metabolic dysregulation and genetic variations in mood disorders. This knowledge could be used to filter out the associated individual parameters to MDD pathophysiology, understand the causal mechanism and develop biomarker panels that aim to profile a diverse array of hormones, cytokines, inflammatory, metabolic, and genetic markers.
Our aim is to look at all the available biological parameters along with demographic and clinical factors that are associated with MDD risk as well as treatment response. Firstly, we will start with global screening from all the available literature to create a cumulative list of all the significantly associated parameters. Next, we will look for information about these parameters from the patient data retrieved from large biobanks such as UK Biobank. Utilising the information, we will create a prediction model that aims to predict disease risk as well as treatment outcome. Lastly, we will validate the developed model using MDD samples from other populations to assess the predictive accuracy, reproducibility and generalisability of the prediction model.
Developing a predictive model for disease risk and treatment outcomes using a data-driven approach represents a significant advancement. This model could address the existing treatment gap and improve overall response rates in MDD patients. The use of a global population in the meta-analysis and the inclusion of large biobanks enhance the generalizability of the developed model. This global perspective contributes to a more inclusive understanding of MDD across diverse populations.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-contemporary-assessment-of-high-penetrance-melanoma-genes

A CONTEMPORARY ASSESSMENT OF HIGH-PENETRANCE MELANOMA GENES

Last updated:
ID:
52303
Start date:
7 August 2020
Project status:
Current
Principal investigator:
Dr Peter A Kanetsky
Lead institution:
Moffitt Cancer Center, United States of America

Our current understanding of spectrum of disease outcomes associated with inherited genetic variation in major genes, including ACD, CDKN2A, CDK4, MITF, POT1, SLC45A2, TERF2IP, and TERT, that result in familial melanoma is limited. We plan to use genotype and whole-exome sequencing data available in the UKBB resource to agnostically determine associations with other (non-melanoma) cancers and non-cancer phenotypes through linkage to cancer registry, hospital, and primary care files. Results will be combined with those arising from parallel analyses in an independent large US data resource to identify top ranking genetically-associated cancers and non-cancer conditions. Findings will be used to inform follow up of melanoma-prone families across the globe.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-convolutional-neural-network-classifier-for-gender-classification

A convolutional neural network classifier for gender classification

Last updated:
ID:
40807
Start date:
27 September 2018
Project status:
Closed
Principal investigator:
Dr Chao-Gan Yan
Lead institution:
Chinese Academy of Sciences, China

Mental illness cause the heaviest economic pressures to the society among all types of diseases and the patients suffer enormous pain. But the diagnose of the psychiatric disorders is merely based on the subjective diagnose of psychiatrists. Researchers want to develop an diagnose technique which is more objective and accuracy using MRI which is entirely harmless to the patients. One outstanding technique is deep-learning algorithm which is a kind of artificial intelligence (AI) method. We want to distinguish the patients and normal people using the deep-learning algorithm built on MRI. But the proper parameters of the deep-learning model are largely undetermined, so build a deep-learning model for distinguish mental illness patients is much infeasible. As physiology gender is a much robust characteristic of human, which is also an dichotomous variable. We want to built the deep-learning algorithm for gender classification. And then, We would try to transfer the model on mental illness. The project duration is about 12 mouth including data accumulation, preprocessing, building deep-learning model and testing the model.
The present work aims to solve some fundamental necessities for the clinical diagnosis of mental illness, and it may facilitate the development of treatment for mental illness and reduce the economic pressures of society and the suffering of patients.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-cross-sectional-exploration-of-the-factors-contributing-to-the-development-of-sarcopenia

A cross-sectional exploration of the factors contributing to the development of sarcopenia

Last updated:
ID:
11058
Start date:
11 May 2015
Project status:
Closed
Principal investigator:
Professor Ailsa Welch
Lead institution:
University of East Anglia, Great Britain

Sarcopenia is the progressive loss of skeletal muscle mass, strength and function with aging, leading to disability and frailty.

This project aims to explore three questions in cross-sectional analyses:

1. What is the prevalence of low skeletal muscle mass (fat-free mass) and muscle strength in the population?

2. What is the body composition and grip strength of the population and what lifestyle activities and disease conditions might influence fat free mass and strength?

3. What dietary factors influence low fat free mass and muscle strength? This research aims to improve our understanding of the factors leading to the development of sarcopenia which are poorly understood. The Biobank data offers the opportunity to investigate this issue. By understanding the prevalence, as well as lifestyle, disease and nutritional factors that relate to loss of skeletal muscle mass and strength, we will be better able to prevent and treat sarcopenia. This fulfils the purpose of the UK Biobank which is ?to support a diverse range of research intended to improve the prevention, diagnosis and treatment of illness, and the promotion of health throughout society?. The researchers will use baseline data from Biobank to explore the 3 questions outlined above. We plan to understand the prevalence of low skeletal muscle mass (as fat-free mass) and strength in the population. We also plan to find out what lifestyle, diet and other factors might affect fat free mass and strength. Will use standard statistical analysis methods to do this work. The researchers will be based at the University of East Anglia and Plymouth University, Plymouth. We plan to investigate questions 1 and 2 about low skeletal muscle mass (fat-free mass) muscle strength on the full cohort.

For question 3, relating diet to fat-free mass and muscle strength, we plan to use the subset with dietary information available.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-cross-trait-integration-study-of-the-genetic-variants-for-cancer

A cross-trait integration study of the genetic variants for cancer

Last updated:
ID:
83445
Start date:
9 March 2022
Project status:
Current
Principal investigator:
Dr Sipeng Shen
Lead institution:
Nanjing Medical University, China

To date, although lots of common genetic variants (Single Nucleotide Polymorphisms, SNP) have been identified from genome-wide association studies (GWAS), the role of rare variants (variants present in less than 1% of the individuals are coined) in genome sequencing technology remains unknown in cancer. In addition, multiple diseases share the similar cause genetic variants or genes, such as chronic obstructive pulmonary disease, emphysema, and lung cancer. Identification of them may help the early detection disease and precision prevention.
We will conduct a cross-trait integration study including multiple diseases on the UK Biobank samples to identify the rare and common genetic variants associated with cancer and cancer related traits. We will explore the key genes in different diseases and identify the share genes. Further, we will use bioinformatics algorithm to benchmark the genes which are important for the diseases.
In summary, this project may help identify the genetic background of multiple cancers and cancer related diseases. Key genetic variants and genes will be screened out. The total period is 36 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-data-driven-approach-to-identify-clusters-of-alzheimers-and-vascular-dementia-patients-using-electronic-health-records-brain-imaging-frailty-index-and-polygenic-risk-score

A data-driven approach to identify clusters of Alzheimer’s and vascular dementia patients using electronic health records, brain imaging, frailty index, and polygenic risk score

Last updated:
ID:
98554
Start date:
3 July 2023
Project status:
Current
Principal investigator:
Mr Jack Quach
Lead institution:
Dalhousie University, Canada

The aim of our research is to use advanced computer techniques to identify different types of Alzheimer’s disease and vascular dementia based on factors such as frailty, brain health, and genetics. By grouping patients with similar characteristics, we can better understand the unique needs and characteristics of each type of dementia and develop more effective treatment plans. If we can better understand the differences in frailty levels in Alzheimer’s disease and vascular dementia types, it will support the idea that frailty should be considered when diagnosing and treating these conditions. Overall, this year-long research study has the potential to improve our understanding of dementia and improve how it is diagnosed and treated.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-data-mining-based-workbench-advancing-precision-medicine-by-the-use-of-machine-learning-and-expert-knowledge

A Data Mining-based Workbench: Advancing Precision Medicine by the Use of Machine Learning and Expert Knowledge

Last updated:
ID:
44972
Start date:
4 March 2020
Project status:
Closed
Principal investigator:
Professor Folkert Asselbergs
Lead institution:
University Medical Center Utrecht, Netherlands

Precision medicine is a form of healthcare where disease prevention and treatment is tailored to the individual patient. Besides environmental factors and lifestyle, also genetic variation is taken into account. This proposal will aim to simulate potential effects of pharmacological and lifestyle interventions in an individual person. Much of the knowledge we possess about genomic risk factors comes from statistical measures of association from large-scale population studies. The conceptual and practical disconnect between the populations we study and the individuals we want to treat is a major topic in research. The primary goal of this proposal is to develop a methodology based on machine learning to facilitate precision medicine for CVD patients by connecting population and individual genomic phenomena. We aim to develop a so-called data mining-based workbench, which will allow clinicians to carry out thought experiments about the treatment of individual patients using models of CVD risk derived from population-level studies. This will help clinicians understand how these risk factors might be useful for the diagnosis and treatment of an individual, accelerating the translation of genomic findings into the clinic.
The proposed APM-GDM is based on representation learning which means it can be fed with raw data and automatically extract necessary representation for predictions. An ensemble method or a DL network can provide representations at different levels. In neural networks, for example, the output of each of hidden layers is considered as the representation at that level. The higher layers the data belong, the more abstract representations we get for these data. In different studies, these higher-level representations of raw data prove to be very effective for classification or detection problems.
The project will be conducted for three years and intend to use as many individuals as available to satisfy the need for statistical and machine learning.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-deep-analysis-of-genetic-and-phenotype-data-in-common-diseases-across-ethnicities

A deep analysis of genetic and phenotype data in common diseases across ethnicities

Last updated:
ID:
108924
Start date:
11 January 2024
Project status:
Current
Principal investigator:
Dr Ramesh Menon
Lead institution:
MedGenome Labs Pvt Ltd., India

Genome-wide association studies (GWAS) are useful in identifying genetic variants associated with human diseases. A polygenic risk score (PRS) uses this the results from GWAS analysis for assigning a risk score for an individual. The genetic risk only one part of the story. Various clinical/biochemical parameters as well as life-style and food habits contribute to the risk of developing the common disease. By integrating the genetic and non-genetic factors we will be able to get the absolute risk score for a disease and one can monitor how the risk for getting a disease increases or decreases by changing the life-style, food habits or through medications. This can be very useful because the genetic risk cannot be altered (in general), however lifestyle modifications ill alter the risk of getting a common disease such as type 2 diabetes or coronary artery disease. This is the next step towards preventive wellness using genetics. This new knowledge will lead to the development of new diagnosis, prevention and hopefully the identification of medicinal drugs, and improve the public health.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-deep-learning-based-approach-for-phenome-wide-association-studies

A deep learning based approach for phenome-wide association studies

Last updated:
ID:
100316
Start date:
5 April 2023
Project status:
Current
Principal investigator:
Dr Gamze Gursoy
Lead institution:
New York Genome Center, United States of America

The goal of this research is to use deep learning techniques to identify connections between a person’s genetic makeup (genotypes) and their physical characteristics or diseases (phenotypes) using data from the UK Biobank. To do this, our aim is to create a new methodology and to identify specific non-coding genotypes (parts of DNA that do not contain instructions for making proteins) that are related to multiple diseases or physical characteristics. After testing this system with UKBB data, we hope to use this system in a manner that would allow us to analyze data from multiple institutions without exchanging information. The rationale is that traditional methods require data from large number of patients; however having large number of samples in single institutions is not possible, however data is sensitive and therefore cannot be shared across institutions. We plan to test this system using a large number of samples from the UK Biobank. The developed tool will perform phenome-wide association study (PheWAS), which is a powerful way for identifying the relationship between the genetic and a wide range of human diseases and traits. By studying large populations, PheWAS can identify genetic risk factors that may be missed by traditional disease-specific studies. This project will require approximately 3 years. The public health implications of PheWAS studies include a better understanding of the underlying causes of many diseases and the identification of new targets for prevention and treatment. Additionally, PheWAS studies can help to improve the accuracy of diagnosis and the development of more effective treatments. Furthermore, PheWAS results can also help to identify population subgroups that are at a higher risk of certain diseases, which can inform public health interventions and improve health outcomes for those populations. In summary, PheWAS studies have the potential to improve our understanding of the genetic basis of disease, and can inform the development of new treatments and public health interventions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-deep-learning-based-phenome-wide-association-study-for-cardiovascular-disease-and-causal-effects-analysis-between-traits-by-mendelian-randomization

A deep-learning-based phenome-wide association study for cardiovascular disease and causal effects analysis between traits by Mendelian randomization

Last updated:
ID:
198575
Start date:
14 April 2025
Project status:
Current
Principal investigator:
Dr Gan Qiao
Lead institution:
Southwest Medical University, China

This project aims to understand more about Cardiovascular Diseases (CVDs) and their connections to other health conditions. Using advanced deep learning technology, we’ll investigate the relationships between CVDs and various traits, such as hypertension, diabetes, and obesity. Our goal is to prioritize these connections and identify potential genetic causes. Additionally, we’ll use a method called Mendelian randomization to figure out if these traits might actually cause CVDs. By comparing our deep learning approach with traditional methods, we hope to discover new biomarkers and crucial genes related to CVDs (including disease risk and prognosis) in the UK Biobank data.

Coronary heart disease is a complex condition influenced by genes, environment, and lifestyle. Previous studies have identified many genetic factors linked to CVDs, but we aim to go further. Our new deep learning method, DSpaLaRefiner, allows us to analyze the data more effectively, identifying patterns and relationships between different traits. Unlike traditional methods, our approach does not rely on external data, giving us a unique advantage in discovering new insights. By understanding the genetic correlations and disease susceptibilities and prognosis, we hope to find more accurate ways to predict, prevent, and manage CVDs.

We will use the GATK software for quality control and Plink1.9, ‘survival’ R package, and custom R codes for genome-wide association analysis on the UK Biobank data. DSpaLaRefiner, our in-house deep learning algorithm, will play a crucial role in identifying disease-associated variants and understanding the relationships between different traits. Mendelian randomization analysis will help us investigate if there are causal links between CVDs and other health conditions. This comprehensive approach aims to uncover new insights into CVDs and their connections, potentially revolutionizing our understanding of this complex disease.

The duration of this project is expected to be 3 years; we will consider extending the research duration based on the progress or discoveries made during the project.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-deep-learning-based-self-super-resolution-method-for-cardiac-mri

A deep learning based self super-resolution method for Cardiac MRI

Last updated:
ID:
57400
Start date:
9 June 2020
Project status:
Closed
Principal investigator:
Mrs Muzi Guo
Lead institution:
Shenzhen Institutes of Advanced Technology, China

High resolution (HR) Cardiac magnetic resonance images (CMRI) provide more anatomical details and enable more precise analyses, and are therefore highly desired in clinical and research applications. However, acquiring such data with an adequate resolution is time consuming. A common way to partly achieve this goal is to acquire MR images with good in-plane resolution and poor through-plane resolution to save scanning time.The aims of the research project is to improve through-plane resolution when saving MR scanning time.Using super-resolution method based on deep learning could use the mapping between the high in-plane resolution images and simulated lower resolution images, to estimate high resolution through-plane images.This project duration will last 24 months.The expected value of the research would produce high through-plane resolution as well as saving Cardiac MRI scanning time.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-deep-learning-model-for-early-detection-of-neurodegenerative-disorders-like-alzheimers-disease-using-retinal-images

A DEEP LEARNING MODEL FOR EARLY DETECTION OF NEURODEGENERATIVE DISORDERS LIKE ALZHEIMERS DISEASE USING RETINAL IMAGES

Last updated:
ID:
106036
Start date:
4 December 2024
Project status:
Current
Principal investigator:
Mr ANSAR A
Lead institution:
Indian Institute of Information Technology, Kottayam, India

The main aim is to create a deep learning model based on retinal pictures that can distinguish between people with Alzheimer’s disease and people without dementia with a level of accuracy that is consistent across time.
The most widespread type of dementia is Alzheimer’s disease (AD) and It is a brain condition that gradually robs people of their memory, thinking abilities, and ultimately their capacity to complete even the most basic tasks. Alzheimer’s disease (AD) is a complex neurodegenerative condition with a number of known and improbable causes, as well as a wide range of anatomical features. The retina, which is a part of the central nervous system (CNS), has been called a “window to the brain” and a brand-new indicator of Alzheimer’s disease (AD). Retina tests are appropriate for large-scale population screening and research into preclinical AD because of their low cost, simplicity of access, and non-invasive properties. Additionally, a number of cutting-edge techniques for retinal imaging, such as optical coherence tomography (OCT), have been developed that enable the visualisation of retinal alterations at a very fine resolution.

Therefore, the primary goal is to create a deep learning model based on retinal images to identify people who have Alzheimer’s disease and to consistently perform accurately in differentiating between people who have Alzheimer’s disease and people who do not have dementia with respect to both accuracy and sensitivity.

The Project can be completed within 36 months.

So the public can perform the Alzheimers screening at very low expense or even at free of cost and within short while. At present there is no such facility is available for this purpose in most of the developing countries like India. Right now the public have pay a lot of money for Alzheimers screening (for PET Scan or Brain imaging etc).


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-deep-learning-model-for-the-classification-of-cardiac-amyloidosis-among-patients-with-left-ventricular-hypertrophy

A Deep Learning model for the classification of cardiac amyloidosis among patients with Left Ventricular Hypertrophy.

Last updated:
ID:
96272
Start date:
4 April 2023
Project status:
Current
Principal investigator:
Dr Yitschak Biton
Lead institution:
Hadassah Medical Center, Israel

Our research project – “A Deep Learning model for the early detection of cardiac amyloidosis among patients with Left Ventricular Hypertrophy”, aims to develop a deep learning based algorithm for classification of various diseases that cause Left Ventricular Hypertrophy – with an emphasis on cardiac amyloidosis among those diseases.

Left Ventricular Hypertrophy is a state in which the heart muscle (myocardium) is over-thickened, and as a result it’s filling capacity is reduced.
The above described state can arise from a number of diseases, one of them is cardiac amyloidosis.
Cardiac amyloidosis is an underdiagnosed, potentially reversible disease, that cause ‘Restrictive Cardiomyopathy’ which leads to progressive Heart Failure with life threatening arrhythmias. The disease arise from abnormal deposition of proteins in a number of tissues, including the heart.

As for today, diagnosis of cardiac amyloidosis is given after a series of tests, including imaging studies, blood tests, scintigraphy and heart biopsy. All of the above makes the journey of the patient towards diagnosis long, expensive and filled with uncertainty.
Given the underdiagnosis and significant morbidity of cardiac amyloidosis, combined with the availability of treatment with disease-modyfing agents, highlights the importance of early diagnosis and treatment.

Several studies has been conducted in order to built a prediction tool for cardiac amyloidosis. Most of them were based on clinical data alone.
We strongly believe that by applying computational learning methods, We’ll be able to built a robust classification tool, that we’ll allow us to predict cardiac amyloidosis as early as possible, and to start disease modifying treatment before the development of progressive heart failure.

In order to do so, we wish to conduct a retrospective study on UK Biobank patients diagnosed with Left Ventricular Hypertrophy based on echocardiographic/electrogram criteria, in order to generate a prediction model for classification of cardiac amyloidosis. Such classification model will enable us to diagnose the disease at an early state, and provide disease-modyfing medication for slowing its progression.
The requested data is a set of several clinical evaluation tools for the patient with heart disease – cardiac MRI (Magnetic Resonance Imaging), Echocardiography, Electrogram and clinical data.
Apllying image processing computational methods on the above will allow us to extract meaningful features for prediction. Combining those with clinical data, will enable us to train state of the art Artificial Intelligence models based on neural networks, that hopefully will be able to classify correctly and thus predict, cardiac amyloidosis in patients with Left Ventricular Hypertrophy.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-diseases-prediction-model-based-on-common-variants-polygenic-risk-score-and-rare-variants-variant-weighted-gene-burden-scores-in-whole-exome-sequencing-data

A diseases prediction model based on common variants (polygenic risk score) and rare variants (variant weighted gene burden scores) in whole exome sequencing data

Last updated:
ID:
107825
Start date:
27 September 2023
Project status:
Current
Principal investigator:
Dr Yuhuan Meng
Lead institution:
Guangzhou KingMed Transformative Medicine Institute Co. Ltd., China

Our research aims to develop an advanced disease prediction model for complex diseases by integrating both common and rare genetic variants identified through whole-exome sequencing (WES) data. One of the primary objectives of our study is to evaluate the heritability of complex diseases using WES data, shedding light on the underlying genetic factors contributing to disease susceptibility. Additionally, we strive to enhance the prediction accuracy of existing models based solely on polygenic risk scores (PRS). By incorporating rare variants into the model, we hope to capture the additional sources of genetic variation that are often overlooked when focusing solely on common variants.

Previous large-scale population studies have shown that PRS, which consider the effects of common genetic variants, can effectively identify individuals at high risk for complex diseases. However, it is important to note that the overall prediction accuracy of PRS models varies, with most falling within the AUC range of 0.6-0.75. This discrepancy may arise from the fact that most current PRS methods solely rely on common variants and do not fully consider the impact of rare variants on disease susceptibility. Despite their lower frequency, rare variants can possess a substantial effect size and contribute significantly to the development of common diseases, even in individuals with low PRS. By integrating both common and rare variants into our prediction model, we aim to provide a more comprehensive understanding of genetic predisposition.

Our proposed research will leverage the extensive whole-exome sequencing data available from the UK Biobank to identify genetic susceptibility loci associated with complex diseases and develop a prediction model based on WES data. This model will hold great potential to advance our knowledge of genetic risk factors, empowering medical professionals to make informed decisions regarding personalized prevention strategies, clinical interventions, and disease management. By considering both common and rare variants, we hope to gain deeper insights into disease risk prediction and contribute to the broader field of personalized medicine. Ultimately, our research endeavors to reduce disease burden, enhance public health outcomes, and drive evidence-based healthcare interventions tailored to individual genetic profiles.

project duration: 36 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-dynamic-network-approach-for-the-study-of-human-phenotypes-using-uk-biobank

A Dynamic Network Approach for the Study of Human Phenotypes using UK Biobank

Last updated:
ID:
76517
Start date:
3 December 2021
Project status:
Current
Principal investigator:
Dr Fang Fang
Lead institution:
Karolinska Institutet, Sweden

Diseases could be viewed as specific sets of phenotypes affecting one or several physiological systems. It has been shown that diseases whose components link to each other at the cellular level show higher comorbidity in the population. Systematically studying entire sets of comorbidities offer a complementary perspective of disease biology from traditional approaches. The overall aim of this project is to systematically study the associations between several diseases in the human population by building phenotypic disease networks (PDNs), with a focus on the known, strong comorbidities between 1) cardiovascular disease and psychiatric disorder, 2) cancer and psychiatric disorder, and 3) rare but complex diseases such as neurodegenerative diseases and psychiatric disorder. Importantly, we will contrast PDNs estimated in the UK Biobank with PDNs estimated using the entire Swedish population in our parallel study using the Swedish national registers.

Exploring comorbidities from a network perspective could help determine whether differences in the comorbidity patterns indicate differences in genetic background, biological processes, environmental factors, or health care quality provided for each population. This project together with our parallel study using the entire Swedish population will offer unique insights into the structure and mechanisms underlying comorbidities in different human populations and will potentially promote the prevention, diagnosis and treatment of diseases and the overall health throughout the society.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-eye-brain-4-dementia-ai-and-in-silico-models-in-mild-cognitive-impairment-and-dementia-and-beyond

A-Eye-Brain-4-Dementia: AI and in silico models in mild cognitive impairment and dementia and beyond

Last updated:
ID:
712383
Start date:
25 July 2025
Project status:
Current
Principal investigator:
Dr Uazman Alam
Lead institution:
University of Liverpool, Great Britain

Rationale
Dementia is a progressive neurodegenerative disease in which patients encounter frequent delays in diagnosis, leading to increased morbidity. There is a major need of biomarkers for the early prediction as acknowledged by the Alzheimer’s Drug Discovery Foundation. The eye is often seen as the ‘window to the brain’ with much effort dedicated recently to predicting disease such as mild cognitive impairment (MCI) and dementia through images of the retinal microcirculation. The eye is closely coupled to the brain through its circulation, with the eye being perfused through a branch off the internal carotid artery that then continues on to the brain. Furthermore, the eye is coupled to the brain neuronally through projection of the CNS into the retina (neuroretina). As such, it is believed that retinal imaging can be used to monitor changes in the brain which has been demonstrated by recent published data. However, whilst the eye is easily imageable down to the micro-scale, the skull around the brain makes imaging much more difficult. Computational models, on the other hand, can simulate the brain circulation and pressure so we know exactly what is happening in the brain in silico. Whilst utilising artificial intelligence (AI) models retinal imaging can help in the diagnosis and prediction of MCI and dementia.

Aim: To develop a model of the circulation in the brain coupled to the eye, observing retinal circulatory alterations with concomitant change in the brain and development of AI models for diagnosis (detection) and prediction of future disease.

Utilising data in the UK Biobank, his work will entail:
1. Coupling circulation models of the brain and eye (In Silico/digital twin model)
2. Synthetic imaging of the eye and brain pipeline simulation (In Silico/digital twin model)
3. Development of AI model of MCI/dementia detection and prediction of dementia and other diseases


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-foundation-model-for-brain-eye-connection

A foundation model for brain-eye connection

Last updated:
ID:
946023
Start date:
30 October 2025
Project status:
Current
Principal investigator:
Professor Yixuan Yuan
Lead institution:
Chinese University of Hong Kong, China

This project aims to investigate how large-scale multimodal data, including fMRI, DTI, T1w/T2w MRI, fundus photography, OCT, and genetic profiles, can be integrated to reveal common biomarkers underlying both neurological and ocular diseases. It further explores whether a foundation model pre-trained on these data modalities can reliably detect early pathological changes in diseases such as Alzheimer’s, Parkinson’s, strokes, cognitive decline, and retinal degenerations, and designs interpretability strategies to elucidate the pathogenic mechanisms shared between brain and eye, potentially informing new therapeutic targets.

The primary objective is to develop a unified deep learning foundation model capable of processing multimodal brain and eye data, leveraging large-scale UKB data for self-supervised pretraining. The model will be evaluated for its performance in early-stage detection and more accurate diagnosis of neurological and ocular diseases, including Alzheimer’s, Parkinson’s, dementia, stroke, schizophrenia, glaucoma, and hereditary retinal disorders, as compared to single-modality baselines. Additionally, the research aims to incorporate explainable AI tools to identify and interpret the critical imaging and genomic features driving disease risk and progression, and to investigate gene-environment interactions by integrating genetic data with imaging for biomarker and therapeutic target discovery.

The scientific rationale lies in the shared developmental origins of retinal and cerebral tissues, which position ocular imaging as a non-invasive proxy for brain health. Recent studies have demonstrated strong correlations between retinal changes and structural or functional brain abnormalities, yet current approaches rarely exploit the full potential of multimodal data integration. The use of interpretable AI will further enable the linking of model predictions with underlying pathophysiology, guiding early interventions and advancing precision medicine.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-framework-for-analyzing-associations-and-interactions-of-genetic-and-non-genetic-risk-factors-toward-complex-disease-outcomes

A framework for analyzing associations and interactions of genetic and non-genetic risk factors toward complex disease outcomes

Last updated:
ID:
83974
Start date:
18 March 2022
Project status:
Current
Principal investigator:
Dr Hongxi Yang
Lead institution:
Tianjin Medical University, China

Complex diseases result from a combination of genetic, environmental, and lifestyle factors. The vast majority of non-communicable diseases are also referred to as complex diseases, including cardiovascular diseases, cancers, diabetes, Alzheimer’s disease, asthma, and many more, and they are responsible for most of the burden on the health care system. Environmental factors acting alone or in concert with genetic or other host susceptibility factors have long been implicated as major contributors to disease burden. Identifying and understanding factors that influence complex diseases is important to guide health-related choices and medical interventions. Yet identification of specific genetic or environmental factors, their interactions, and their effects on human complex disease has remained elusive. An environment-wide association study (EWAS) is a type of epidemiological study analogous to the genome-wide association study (GWAS). The EWAS can systematically examines the association between a complex disease and multiple individual environmental factors. This research aims to search for genetic and environmental factors associated with complex diseases on a broad scale by performing GWAS and EWAS. We also plan to develop a framework for quantifying the contribution of genes, environmental factors, and gene-environment interactions to complex diseases, which can help understand the role of genetics and environment in determining the course of a disease. This project will enhance our understanding of the genetic architecture and pathways of complex diseases. These results generated from this project could inform public health authorities and the general public themselves which risk factors associated with complex diseases should be paid more attention to and the extent to which the changes of environmental and lifestyle factors may help offset the genetic risk of complex disease. This project is excepted to take around 36 months to complete.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-framework-to-assess-causal-effects-in-observational-data-through-deep-digital-phenotyping-and-creating-digital-twins

A framework to assess causal effects in observational data through deep digital phenotyping and creating digital twins

Last updated:
ID:
71033
Start date:
17 May 2021
Project status:
Current
Principal investigator:
Dr Rohan Khera
Lead institution:
Yale University, United States of America

In a real-world setting, we have been limited in our ability to infer whether a particular lifestyle or a treatment received by an individual leads to a good or a bad health outcome, despite systems to measure both these lifestyle features and treatments, and the outcomes. A major limitation is that inferring such a cause-and-effect association requires that at least identical individuals exist, which differ only on the lifestyle or treatment in question. The ability to define individuals that resemble each other has, however, been limited by our ability to actually leverage only crude characteristics even though electronic health records as well as electrocardiographic and imaging data may capture some of these distinct patient features.

The current proposal investigates a novel strategy to create an efficient way to define each individual using their data captured in all these high-quality data sources. This virtual representation of each person (referred to as their “digital twin”) will be used to identify other individuals who resemble this person on a set of measurable characteristics.

Our work will evaluate the least amount of unique data sources that are required to define digital twins. Pairs of digital twins will be followed over time for the development of adverse cardiovascular events while focusing on identifying lifestyle or treatment differences that may underlie differences in the trajectory of outcomes among such digital twins.

Collectively, our investigations will leverage the uniquely powerful contributions of UK Biobank participants to make methodologic advances that allow us to gain deeper insights from observational studies, potentially expanding their role in scientific discovery.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-functional-genomics-approach-to-investigate-the-molecular-bases-of-rare-genetic-diseases

A functional genomics approach to investigate the molecular bases of rare genetic diseases

Last updated:
ID:
88515
Start date:
29 September 2022
Project status:
Current
Principal investigator:
Mr Lorenzo Vaccaro
Lead institution:
Telethon Institute of Genetics and Medicine, Italy

The rising of the post-genomic era opened new perspectives for treatment opportunities for many diseases, by allowing researchers and clinicians to focus on the molecular bases of such diseases in an easier fashion. Unfortunately, for most of the genetic disorders, this process is hampered by the limited cohort of patients that limits the knowledge about the driver molecular functions altered by such mutations. This, in turn, leads to an inadequate treatment of patients and to an inefficient healthcare system. In the last years, many researchers tried to build comprehensive tools that have the advantage to lead to a high number of information about the mechanisms of diseases. On the other side, it is very difficult to find a good balance between the cost of the screening and the amount of generated data. In our laboratory, we developed a method to efficiently screen thousands of protein variants in a single multiplexed experiment and with a moderate cost. As a proof of principle, we applied it to P63, a transcription factor that plays a key role in skin development and whose mutations are causative of genetic disorders. To validate the method we aim to identify patients with novel variants that we identified with our screening and “sane” patients that carry potentially pathogenic mutations. To solve this task, UKBiobank will be fundamental, by allowing us to access the genetic data of thousands of individuals with no hard disease-related phenotypes. If confirmed, our screening method could represent a milestone in the field of genetic disorder, by providing the clinicians a key weapon to fasten a specific diagnosis for a disease and, in turn, to adequately take care of the patient with a specific treatment


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-fusion-study-of-mendelian-randomization-genomics-and-radiomics-on-intracranial-arteriosclerosis

A Fusion Study of Mendelian Randomization, Genomics, and Radiomics on Intracranial Arteriosclerosis

Last updated:
ID:
815678
Start date:
3 July 2025
Project status:
Current
Principal investigator:
Dr Xiaoming Ma
Lead institution:
Suzhou Hospital, Affiliate Hospital of the Medical School of Nanjing University, China

At present, there are already sufficient studies on peripheral arterial disease, but few studies have focused on whether there are pathogenic gene mutations or abnormal protein function in intracranial arteriosclerosis. Therefore, this study hopes to conduct co-localization analysis of genomics and proteomics, use Mendelian randomization methods for causal inference, and use imaging data for fusion analysis in the hope of discovering the characteristics that lead to intracranial arteriosclerosis and premature intracranial arteriosclerosis.

Intracranial Atherosclerosis (41202, ICD Code: I67.2) is a major contributor to ischemic stroke and cognitive decline, especially in Asian populations. Despite its clinical significance, current research predominantly focuses on extracranial or peripheral arterial disease, leaving the pathogenesis of intracranial arteriosclerosis insufficiently understood. Unlike peripheral atherosclerosis, intracranial arteriosclerosis presents distinct biological behavior and risk profiles, suggesting that different molecular mechanisms may be involved.

Recent advances in genomics and radiomics have enabled multi-layered exploration of complex diseases. However, these modalities are rarely integrated in a unified analytic framework to investigate intracranial vascular pathology. In particular, the genetic architecture underlying intracranial arteriosclerosis remains poorly defined, and it is unclear whether certain protein-level changes mediate the effects of inherited genetic variants. Furthermore, the relationship between these molecular signatures and radiological phenotypes of intracranial artery disease has not been systematically studied.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-generative-model-for-brain-age-predictions

A generative model for brain-age predictions

Last updated:
ID:
65657
Start date:
9 November 2020
Project status:
Closed
Principal investigator:
Dr Chiara Mauri
Lead institution:
Technical University of Denmark, Denmark

The goal of this research is to develop a generative model for predicting age of healthy subjects based on their T1-weighted brain scans, in particular from grey matter segmentation maps. The predictive method entails a forward model that uses the target variable (age) and other demographic variables to predict the grey matter image of
a subject. In a subsequent step, the model needs to be “inverted”, to obtain the prediction of age, based on the subject’s grey matter segmentation map and some demographic variables. T1-weighted scans of healthy subjects from the UK Biobank will be processed to obtain grey matter segmentation maps, and then used to train and test the predictive method. The prediction performances achieved by the proposed method will be compared with the ones of current state-of-the-art methods for brain age prediction. The age prediction task is considered as a way to test the proposed predictive method, which can be used later on for prediction tasks in a clinical setting, such as prediction of some clinical scores in Multiple Sclerosis. Furthermore, brain age prediction is of clinical interest itself: several studies have shown that the difference between real age and predicted brain age correlates with some measures of disability. Therefore this “brain age gap” can be used as a biomarker, to study healthy ageing and to characterise pathological deviations underlying several diseases. The expected duration of the project is 2 years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-generative-modeling-for-normative-aging-brain-mris-and-neurological-abnormality-detection

A generative modeling for normative aging brain MRIs and neurological abnormality detection

Last updated:
ID:
106908
Start date:
31 August 2023
Project status:
Current
Principal investigator:
Dr Youngjin Yoo
Lead institution:
Siemens Medical Solutions USA, Inc, United States of America

Human brain changes with normal aging and incidence of brain diseases also rise with age. In order to provide aging people with medical treatments at the right timing, expert routine inspection of brain structure and function in medical images, which is an important proxy to examine brain changes with increasing age, is important for determining if the changes are normal or not. However, this expert inspection can be very time-consuming and expensive. There have been researchers’ efforts to automate this manual inspection of brain MR images using computer algorithms, but it has been hampered by high computing demand and limited amount of brain MR image datasets which are crucial for developing accurate automated computer algorithms. Due to the recent advent of super-computers and big public brain MRI datasets such as the UK Biobank data, developing artificial intelligence (AI) based automated tools for examining and creating brain MR images is being rapidly accelerated.
In this study, we propose developing computer algorithms that can automatically generate a healthy brain medical image of a subject, in which its brain appearance is normal for the subject’s specific age. The computer algorithm will automatically create a normal-for-age healthy brain medical image when the subject’s demographic and clinical information and real brain medical image are given to the computer algorithm. Then the difference between the AI-generated healthy brain image and the real brain image will be automatically analyzed to help physicians decide if the subject’s brain condition is healthy or abnormal for the specific age. This appearance gap between real brain MRI image and computer-generated healthy brain MRI image will serve as a medical feature indicating potential risk of having brain diseases and the severity of them. The new AI based computer algorithm would enable to discover new medical indicators that signify potential risk of a subject to develop further aging-related brain diseases.
We will validate the new computer algorithm with both healthy subjects and patients with brain disease to determine if there is any meaningful relation between the brain appearance gap and medical symptoms in patients with brain diseases. The outcome of the study will help improving the accuracy of routine brain MR exams and reducing the associated expert time and cost, which would be beneficial for managing aging population more effectively. The project will be conducted over multiple years as the UK Biobank dataset continues including more new subjects.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-genetic-investigation-of-pleiotropy-using-the-uk-biobank-data

A genetic investigation of pleiotropy using the UK Biobank Data.

Last updated:
ID:
9659
Start date:
1 May 2015
Project status:
Current
Principal investigator:
Dr Vincent Plagnol
Lead institution:
Genomics Ltd, Great Britain

The aim of the proposed research is to understand pleiotropy: that is the nature, extent, and effect of genetic variation on multiple phenotypes. The extent of pleiotropy in humans is an important open question. Apart from inherent interest, it is directly relevant to the use of human genetics for improving drug development pipelines (see below). By their nature, studies of pleiotropy require data on multiple phenotypes. We aim to investigate pleiotropy using genetic data together with baseline measurements and the biomarker data (as it becomes available). Our analysis ultimately aims to inform decisions about which genes and pathways are the best targets for drug development. The efficacy and safety of therapeutics depends on the consequences of perturbations, by the drug, of particular gene products. Genetic variants also perturb the nature or amount of gene products, and is informative for drug efficacy, with effects on other phenotypes informative for on-target safety effects. The proposed work, mainly on non-clinical phenotypes, will involve proof-of-principle studies and development of statistical methods. We will make a further application when more clinical phenotypes are available in UK Biobank The research will use computers to build statistical models of the correlation between the genetic variation in an individual?s genome and biological measurements collected by UK Biobank. We can use the research to ask: if the genetic difference in a gene mimics or is related to the effects of a treatment what is likely to be the (positive and negative) effects of giving it to patients? To do this effectively we will look at the relationship between genetic variation and multiple phenotypes at the same time. Full cohort.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-genetic-study-of-abnormal-fat-deposition-and-its-impact-on-human-diseases

A genetic study of abnormal fat deposition and its impact on human diseases

Last updated:
ID:
71668
Start date:
1 November 2021
Project status:
Closed
Principal investigator:
Dr Peng Chen
Lead institution:
Jilin University, China

Obesity and fatty liver disease are the results of abnormal fat deposition, which means fat tissue accumulated in an excessive amount in a particular body part. They could increase the risk of life-threatening conditions, such as heart attack. In this project, we aimed to add novel knowledge of the genes and environmental factors behind abnormal fat deposition to the existing literature. We will first calculate the fat content in the whole liver using a software developed by our team. After integrating with other fat deposition phenotypes in the UK Biobank, we will try to find the genes associated with fat deposition, as well as the environmental factors that interact with these genes. The relationship between abnormal fat deposition and human diseases or traits will also be investigated.
With our results, the prevalence of fatty liver disease in the UK could be estimated in a much larger cohort. Our results would also shed light on the genetic mechanism of the diseases related to abnormal fat deposition.
The project is expected to last for two years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-genetic-study-of-human-cognitive-function-and-age-related-cognitive-decline

A genetic study of human cognitive function and age-related cognitive decline

Last updated:
ID:
7908
Start date:
1 December 2014
Project status:
Closed
Principal investigator:
Professor Albert Tenesa
Lead institution:
University of Edinburgh, Great Britain

The aim of the current study is to gain further understanding of the genetic factors that underlie human cognition and cognitive decline by identifying the genes that contribute to variation in cognitive function in the UK Biobank.
The project will correlate genetic and phenotypic variation to identify genes that contribute to memory and processing speed (pairs matching, prospective, numeric, light pattern memory, reaction time and fluid intelligence), and develop predictors of cognitive impairment based on genetic markers.
Cognitive impairment is a major health and social issue in ageing populations. Age-related cognitive decline is costly to the individual, his relatives and society in general. It represents a major financial burden to the health services and often precedes dementia, illness or death.
Individual variation in cognitive ageing is partly genetic and partly environmental. About 50% of the cognitive variation is genetic. Identifying the genes that contribute to cognitive ageing would allow developing better prediction models of cognitive impairment, thereby facilitating early intervention; and better understanding of the molecular basis of disease that could eventually provide better or new treatments. Cognitive measurements and their change will be compared to the genetic variations measured from blood DNA. We expect that variation in DNA within, or nearby, genes relevant to cognitive function will correlate with differences in cognitive function among individuals.
We will use statistical methods to test if any of the hundreds of thousands of single nucleotide polymorphisms (SNPs), a type of genetic variation, measured in the UK Biobank is associated with changes in cognition. Subset of the cohort with cognitive measurements that had cognitive measurements at recruitment.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-genome-wide-and-pheno-wide-association-study-of-common-diseases-on-900000-individuals-from-us-and-uk

A genome-wide and Pheno-wide association study of common diseases on 900,000 individuals from US and UK.

Last updated:
ID:
19416
Start date:
10 January 2017
Project status:
Closed
Principal investigator:
Professor Christopher O'Donnell
Lead institution:
VA Boston Healthcare System, United States of America

The US Million Veteran Program (MVP) partners with Veterans to study how genes affect health, by safely collecting blood samples and health information from up to one million Veteran volunteers. So far, four beta projects were approved and funded to study Cardiovascular risk factors, Multi-substance use, pharmacogenomics of kidney disease, and Metabolic conditions. Here, we aim to use the genotype and phenotype data from UK biobank to replicate novel findings from MVP, to become a genetic study with unprecedented power for discovery and replication. The goal of MVP is to better understand how genes affect health and illness in order to improve health care for Veterans. This is well in line with the UK Biobank?s aim to improve the prevention, diagnosis and treatment of a wide range of serious and life-threatening illnesses. The research participants of MVP are US Veterans volunteers, their motivation is to help transform health care, not only for themselves, but for future generations of veterans and the general population. We propose to: (1). Use the MVP data as a discovery for genome-wide and pheno-wide analysis to discover novel associations with the four group of traits mentioned above (with a plan to extend to more traits including cancer and neurodegenerative traits); (2). Follow up the novel findings in the UK Biobank data, where we use the same protocol for statistical analyses; (3). Explore the combination of two largest cohorts (MVP in US and UK Biobank in UK) to further characterize the genetic architecture of complex traits with an unprecedented statistical power. MVP will be the single largest cohort with a rich collection of clinical records over two decades. Also, the MVP used the same Affymetrix Axiom genotype array as UK Biobank, which made synergizing these two studies a natural choice. We are requesting to use the full cohort of UK Biobank.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-genome-wide-and-phenome-wide-association-study-of-daytime-napping

A Genome-Wide and Phenome-Wide Association Study of Daytime Napping

Last updated:
ID:
53562
Start date:
31 March 2020
Project status:
Closed
Principal investigator:
Dr Peng Chen
Lead institution:
Jilin University, China

1. Aims: Disease susceptibility and the contributions of daytime napping to these diseases change along with aging. We aims to capture the trend in which daytime napping affects human health conditions in different age groups.
2. Scientific rationale: The role of daytime napping in maintaining body health was said to be different in children and adults. At the same time, academic investigators did not get a putative perspective whether daytime napping is good or bad for people. To solve the puzzle, we designed a study using UK Biobank genotype and phenotype data stratified by age groups. In this way, we are able to infer the casual relationship between daytime napping and many other human traits, hence, bring up a more comprehensive perspective of the consequences of daytime napping.
3. Project duration: Two years will be needed to complete this project.
4. Public health: A proper guidance of daytime napping (e.g. frequency and duration) depending on age will be proposed.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-genome-wide-association-study-for-occupational-mental-health

A genome-wide association study for occupational mental health

Last updated:
ID:
56010
Start date:
20 November 2019
Project status:
Current
Principal investigator:
Professor Qian Li
Lead institution:
Beijing Foreign Studies University, China

Occupational mental health problems are dramatically increasing during the past few years. It has been very costly to both individuals and the society. Although the past research in social science has explored the varied social and psychological factors that may influence individual’s occupational mental health, the specific genetic variants and neurological foundation responsible for this have largely remained elusive. Therefore, to better understand its biological mechanism and provide with possible career development advice for millions of employees, our research aims to identify the relationship between individual’s genes, brains, and mental health across different occupations. Specifically, we will firstly explore the genome-wide association with employees’ mental health in different occupations, and then further explore the functioning of brains in this relationship. Moreover, we will also investigate the gene × environment interactions on the occupational mental health. We are expected to investigate these research questions in the following three years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-genome-wide-association-study-gwas-of-biological-age-measures-in-uk-biobank-with-a-focus-on-genetic-variations-of-collagens-elastin-and-pro-inflammatory-cytokines

A Genome-Wide Association Study (GWAS) of biological age measures in UK Biobank, with a focus on genetic variations of collagens, elastin and pro-inflammatory cytokines

Last updated:
ID:
48181
Start date:
25 June 2019
Project status:
Closed
Principal investigator:
Professor Roman Romero-Ortuno
Lead institution:
Trinity College Dublin, Ireland

As we age, we accumulate health problems like medical conditions and/or disabilities. However, the speed at which we accumulate those problems is highly variable. On one hand, some people have already accumulated a high number of health problems at a young age, and they look ‘frail’ and/or ‘older than their age’. On the other hand, some people have an advanced age but live with very few health problems, are ‘resilient’ and seem ‘younger than their age’.

The reasons behind this wide variation between age (chronological age) and health problems (biological age) are not well known but there may be genetic reasons. We know that variations in genes that deal with inflammatory reactions in the body have been associated with predisposition to accumulate health problems. However, we also need to consider the environment where those inflammatory reactions take place, which is the so-called ‘connective tissue’. The connective tissue is made of proteins such as collagen and elastin. Collagens are the most abundant proteins in humans, and they provide strength and shape to the tissues. In addition, a lot of chemical reactions (including inflammatory reactions) happen on their surface, and many other cells depend on connective tissue for their normal functions. In addition, collagens and elastin form the ‘building blocks’ of blood vessels, and it is possible that genetic variations in those could have whole-system consequences in terms of resilience or a tendency to accelerated ageing.

We can measure accumulation of health problems using indices of multimorbidity (that is, the number of diagnosed medical conditions) and disability (that is, problems performing activities of daily living); in addition, in UK Biobank there are also validated measures of frailty (such as the frailty index or the frailty phenotype). We propose a Genome-Wide Association Study of these biological age measures in UK Biobank in order to gain a better understanding of the genetic determinants of biological ageing in humans. Understanding the genetic determinants of frailty and resilience in middle-aged and older people could have a significant public health impact.

Our proposed 3-year project will provide an initial phase for a collaboration between the applicant (Prof. Roman Romero-Ortuno: RRO) and Prof. Ross McManus (Professor of Molecular Medicine at Trinity College Dublin). RRO has access to The Irish Longitudinal Study on Ageing (TILDA, http://tilda.tcd.ie) and the proposed pilot work in UK Biobank will inform further collaborative TILDA research looking at novel markers of resilience and successful ageing.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-genome-wide-association-study-in-anca-associated-vasculitis

A Genome-Wide Association Study in ANCA-Associated Vasculitis

Last updated:
ID:
57877
Start date:
16 June 2020
Project status:
Closed
Principal investigator:
Dr Paul Lyons
Lead institution:
University of Cambridge, Great Britain

Vasculitis is a rare disease where the immune system -which normally fights infection- mistakenly attacks the blood vessels and surrounding tissues, leading to dysfunction of potentially any organ and system in the body. ANCA-associated vasculitis (AAV) is a type of vasculitis characterized by the abnormal production of antibodies against inflammatory blood cells called neutrophils. These antibodies, known as ANCAs (Anti-Neutrophil Cytoplasmic Antibodies), are directed against one of two neutrophil components, myloperoxidase (MPO) and proteinase-3 (PR3), and activate neutrophils upon binding, leading to inflammation of small and medium-sized vessels.
Disease manifestations can be very different from one patient to the other, ranging from life-threatening (especially when the kidneys, the lungs or the heart are involved) to a relatively benign presentation. Drugs that suppress the immune response are the mainstay of treatment and are usually effective, but can have severe side effects. What causes the disease, and why it can be so different from one patient to the other, is not well understood.
We previously conducted the first large-scale genetic association study in more than 1200 patients with AAV, and identified genetic variants that differentially predispose to the 2 main subsets of the disease, namely PR3-positive and MPO-positive AAV.
This project aims to further look into the genetic architecture of AAV by studying a new cohort of over 3000 patients, recruited from all over Europe thanks to the European Vasculitis Genetics Consortium (EVGC). The genetic makeup of AAV patients will be compared with healthy controls from the UK BioBank, and the results will be combined with those of our previous study. By substantially increasing the number of patients and healthy controls to study, we will have significantly higher chances to discover new genetic variants that affect predisposition to AAV. Moreover, we will look into whether different genetic profiles can explain clinically relevant differences between patients (for instance, whether the vasculitis affect the kidneys, or the lungs).
A better understanding of the genetic architecture of AAV can provide novel insights into biological processes that play a key role in the disease. This line of research can identify promising drug targets, aiding the development of new medicines with an improved safety profile. Moreover, the discovery of genetic markers that modulate disease course may help doctors to better tailor treatment to patients’ needs, improving disease outcomes and quality of life.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-genome-wide-association-study-in-gout-the-nz-eurogout-us-consortium

A genome-wide association study in gout: the NZ/Eurogout/US Consortium

Last updated:
ID:
12611
Start date:
1 June 2015
Project status:
Closed
Principal investigator:
Professor Tony Merriman
Lead institution:
University of Otago, New Zealand

Gout is a form of arthritis with the primary cause elevated levels of uric acid. In some but not all people the uric acid crystallises in the joints with gout resulting from a painful immune system response. Genetic causes of elevated uric acid are relatively well understood, however knowledge of the genetic factors controlling crystallisation of uric acid and subsequent immune response are extremely poorly understood. Therefore the aim of the proposed research is to identify genetic variants that influence the risk of gout, particularly those controlling crystallisation and immune response. UK Biobank has the aim of improving the prevention, diagnosis and treatment of a wide range of serious and life-threatening illnesses. Gout is a serious illness. It affects up to 3% of adults in Westernised countries and can be a disabling form of arthritis in some people. It is also associated with other serious metabolic conditions such as diabetes and heart and kidney disease. With our aim to find out why some people get gout and others don’t, the proposed purpose directly meets the UK Biobank’s stated purpose. We will create a group of people with gout and a group without gout, matched by age and sex. Gout will be defined as those with doctor diagnosed gout, as determined from the medical records. We will then take the genome-wide genotype data and scan the genome for genetic variants that have a statistically significant difference between people with and without gout. These genetic variants will pinpoint genes involved in gout. To determine which genes are involved in crystallisation and immune response we will also compare to people with elevated uric acid but without gout. All people of European Caucasian ancestry with doctor-diagnosed gout from medical records (10-15,000). Two age- and sex-matched controls for every case (20-30,000). A separate group of asymptomatic hyperuricaemic controls (20-30,000). We would select the participants, therefore we request access to the entire dataset.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-genome-wide-association-study-in-patients-with-intracranial-atherosclerosis

A GENOME WIDE ASSOCIATION STUDY IN PATIENTS WITH INTRACRANIAL ATHEROSCLEROSIS

Last updated:
ID:
51946
Start date:
4 March 2020
Project status:
Current
Principal investigator:
Mr Israel Fernandez-Cadenas
Lead institution:
Institut Recerca Sant Pau, Spain

Large-artery intracranial atherosclerosis (ICAs) is a major cause of stroke worldwide. Different clinical risk factors have been associated with risk of ICAs. However, there is still a gap in the knowledge of the physiopathology of the disease. Furtheremore, there is not any drug to prevent or treat stroke in patients with ICAs.

Genome-Wide Association studies (GWAs) have identified 6 loci associated with large-artery stroke. However, no GWAs has been performed to find genetic variants associated with ICAs. Given the relevance that genetics seems to have in this pathology, our aim is to identify genetic risk factors associated with ICAs using a GWAs approach. This data could help to identify key factors for this disease. These factors could be then targetted by existing or new drugs. Some studies from Astrazeneca has demonstrated that drugs developed using GWAs data have a very higher probability to be used in clinics than drugs developed from animal studies.

We have analyzed 107 patients with ICAs (94.6%symptomatic) and 316 population-based controls and we have replicated the results in two cohorts of 444 and 1,270 stroke patients attributed to intracranial and extracranial atherosclerosis, respectively, and 25,643 controls. We have identified one locus at 18q22 associated with the presence of ICAs. This region was replicated in an independent cohort f (p-value=0.009 for the top SNP1). There was no evidence for association of this polymorphism with extracranial atherosclerosis.
This locus is located near a prostaglandin-reductase gene previously associated with VEGF levels. This protein has been previously related with developement and progression of angiogenesis. Angiogenesis is a kew factor in the progression of patients after suffering a stroke. Further studies are needed to really see the implication of the polymorphisms in angiogenesis.

Now, our aim is to do a GWAs meta-analysis with the UK Biobank data, including all patients with stroke attributed to intracranial stenosis or occlusion in order to increase the sample size and to identify new polymorphisms associated with this disease. Furthermore, the polymophisms identified will be further studied to find biological implications.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-genome-wide-association-study-of-human-transmission-distortion

A genome-wide association study of human transmission distortion

Last updated:
ID:
64662
Start date:
16 September 2020
Project status:
Closed
Principal investigator:
Dr Cory Weller
Lead institution:
National Human Genome Research Institute, United States of America

During reproduction, a parent passes on a single copy of a given gene: either a copy from their mother, or a copy from their father. However, what are referred to as “selfish” genetic elements can stack the deck in their favor, increasing odds of being passed on. While selfish genetic elements are described for a variety of organisms, their evidence in humans has been inconclusive. We aim to test for the presence of such non-randomly inherited genetic elements in the human genome, particularly with respect to variation predisposing a parent to having boys versus girls. Researching such “selfish” elements can inform us of the role genes play with regard to difficulty conceiving, sterility, miscarriage, and developmental disorders.

There are multiple examples in animal systems demonstrating how the inheritance of sex can be biased from the expected 1:1 ratio. One mechanism leading to sex ratio bias can be unequal transmission of sex-determining chromosomes. For example, a man who produces more mature sperm bearing Y chromosomes than X chromosomes would present as a bias toward having sons. Alternatively, sex ratios could be biased due to differences in the survival (or viability) or male or female embryos. A mother passing on an X chromosome mutation that makes it less likely for male embryos to implant or survive through pregnancy, for example, would manifest as a bias toward having daughters. One specific case of reduced viability, the “Mother’s Curse” hypothesis, predicts an accumulation of mitochondrial mutations that only are harmful to males.

We will test for the above biased transmission mechanisms, leveraging the sample size within the UKBiobank. By investigating genetic factors related to biased sex ratios, this project aims to shed light on heritable factors related to sex-specific developmental disorders or sterility. This project should be completed within two years from the date of data acquisition.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-genome-wide-association-study-of-resilience

A Genome-wide association study of resilience

Last updated:
ID:
72298
Start date:
6 December 2021
Project status:
Current
Principal investigator:
Dr Marc Woodbury-Smith
Lead institution:
Newcastle University, Great Britain

It is widely recognised that early childhood trauma (including neglect) is strongly associated with later difficulties during adolescence and adulthood, including mental health problems and poor social and health outcomes. Some people, however, seem to be resilient to early adversity, and despite trauma or neglect during their childhood manage to have a good quality of life in adulthood. We know that psychological characteristics associated with such resilience include positive emotions, optimism and adaptive coping strategies. Other factors, such as good parenting and social support, particularly close, confiding relationships, can engender resilience, particularly through a ‘buffering’ effect on life events and adversity.

Other research has also focussed on biological factors that might influence how resilient a person is. These include hormonal factors as well as the body’s ‘stress response’. Genes are also believed to be important, and by studying the coding regions of key hormonal and stress response genes, some small studies have shown that differences in the coding regions of these genes are associated with how resilient a person is.

As there are probably many genes that are important, looking at the association between variation in a person’s DNA and their resilience should examine all genes and the DNA between genes. This is undertaken using a method called genome-wide association (or GWA), which is the preferred option for studying the genetics of any complex trait. We now plan to study resilience in this way using the UK Biobank, a population-based cohort of ~500,000 participants recruited in the UK. Phenotype information pertaining to early adversity and later adaptation have been collected on participants in addition to their DNA. The aim of this study is to undertake a GWA study of resilience to identify markers (polymorphisms) that are associated with a person’s resilience. In this way, we hope to identify some of the polymorphisms that are associated with resilience as well as the genes that are important and their pathways and functions in the body.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-genome-wide-association-study-of-tinnitus-to-identify-genes-of-interest-and-potential-therapeutic-targets

A Genome-Wide Association Study of Tinnitus to Identify Genes of Interest and Potential Therapeutic Targets

Last updated:
ID:
57266
Start date:
9 March 2020
Project status:
Closed
Principal investigator:
Dr Jian Zuo
Lead institution:
Creighton University, United States of America

Tinnitus is a distressing and persistent condition in a wide portion of the population, but little is known about its functioning and direct cause. Without a solid foundation of understanding, it becomes incredibly difficult to develop effective treatments. In order to develop drug therapies for tinnitus, then, it is critical to understand its mechanism of action. To do this, we plan to conduct a genetic study based on data from both the UK Biobank and the U.S. Department of Veteran Affairs to identify genes impacting the development and severity of tinnitus. Then, by relating these genes back to their corresponding pathways and validating the change in activity of these pathways in between healthy and tinnitus-affected individuals, we can identify potential targets for tinnitus therapies. Using a data-based drug screen, it is possible to identify compounds that change the activity of these targets, and by extension, alter the effects of tinnitus in an individual. We are then able to test these compounds in a mouse model that corresponds to the cause of tinnitus, which may be better representative of the effectiveness of the drug against a known cause of tinnitus.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-genome-wide-study-of-clinically-relevant-g-protein-coupled-receptor-variants

A Genome-Wide Study of clinically relevant G-Protein Coupled Receptor Variants

Last updated:
ID:
72673
Start date:
13 May 2022
Project status:
Current
Principal investigator:
Dr Jens Kleinjung
Lead institution:
Nxera Pharma UK Limited, Great Britain

The project aims at the development of novel medicines for diseases that are related to structures, so-called receptors, which are located on the outside of cells in the human body and that can bind certain substances and thereby convey signals to the inside of the cell. In this project we focus on a particular kind of receptors, the “G-protein coupled receptors” (GPCRs). GPCR receptors are important for conveying signals originating from nerves, hormones, and the immune system, which are all important for keeping the body in a balanced healthy state. The central role of receptors in signalling makes them important drug targets. However, only a minority of GPCR receptors are currently used in medical treatment, because our knowledge about them is incomplete.

In this project, which we estimate to take about three years, we would like to use differences in the genetic information among people, and to link their medical records to identify receptors that could be involved in diseases.

About 60 new changes per generation are added to a person’s genetic material (DNA), and over time many of those changes — called mutations — have also affected GPCR receptors, as these receptors are encoded by the human DNA sequence.

The GPCR mutations that we will see in healthy people in this project will enable us to make modified GPCRs with normal function. The GPCR mutations that turn out to be related to diseases will be engineered to study their modified function.

Most diseases can be traced back to a genetic origin, often involving multiple mutations. By analysing the genetic information of many people, we can determine which mutations are associated with groups of people who are affected by a disease compared with groups of healthy people. GPCR receptors carrying disease-related mutations can be made and tested for their biological function or interaction with potential drug molecules. We intend to study diseases related to inflammation (for instance, gut inflammation) and neurodegeneration, which is the irreversible loss of nerve cells, leading to dementia. Alzheimer’s disease is the most common cause of dementia, and therefore it is important to find treatments to cure it. The relatively large number of people present in the UK Biobank (or their relatives) who are affected by Alzheimer’s disease also means that we can fruitfully use the data collected in the UK Biobank to relate GPCR receptors to neurodegeneration.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-genomewide-association-study-of-autoimmunity

A genomewide association study of autoimmunity

Last updated:
ID:
6728
Start date:
1 May 2015
Project status:
Current
Principal investigator:
Professor Stephen James Sawcer
Lead institution:
University of Cambridge, Great Britain

Autoimmune diseases, such as multiple sclerosis, type 1 diabetes, coeliac disease, rheumatoid arthritis, lupus and thyroid disease, are common health problems in the UK. Comparing and contrasting the results from disease specific GenomeWide Association Studies (GWAS) has shown that there are common aetiological mechanisms underlying these diseases and in this context we are proposing to undertake a GWAS with autoimmunity as the primary endpoint using genotype data from the UK Biobank as controls. Each disease specific group in this collaborative application has genotype data from many thousands of affected UK individuals. Imputation will enable us to compare these existing data with UK Biobank subjects. To supplement these already extensive existing data we will be applying for funding to undertake genotyping of the UK Biobank chip in several thousand previously untested cases. This GWAS would be well powered to identify even low frequency risk variants. The results generated might provide invaluable insight into aetiology and thereby enhance the potential development of safe, effective, rational therapies.
The main analysis would exclude those participants with self-reported autoimmune disease at baseline. Further analyses would be done using self-reported cases and controls from UK Biobank in combination with data from other studies. Disease specific analysis will also be performed with similar treatment of the relevant self reported cases. It is anticipated that most if not all of the untyped UK autoimmune disease patients are not currently part of UK Biobank. Full Cohort


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-genomic-data-science-framework-for-the-1000-arab-genome-project

A Genomic Data Science Framework for the 1000 Arab genome project

Last updated:
ID:
64823
Start date:
1 March 2021
Project status:
Closed
Principal investigator:
Professor Andreas Henschel
Lead institution:
Khalifa University of Science and Technology, United Arab Emirates

Our motivation for studying the UK Biobank data set is based on the observation that traditional detection methods for finding indicators of a particular disease state are not well translated with ethnic minorities. The intention is to develop algorithms that can learn genetic characteristics from large datasets, and then develop transfer learning methods for smaller datasets, ie. Understudied populations. We intend to deal with population specificity, with a particular focus on the population of the UAE.
For benchmark purposes, we aim to study diseases, namely coronary artery disease (CAD), atrial fibrillation, type 2 diabetes, inflammatory bowel disease, and breast cancer. The UK Biobank comprises a dataset from ethnically diverse participants, which is a crucial requirement for our purposes.
In particular, we will first perform population stratification on the dataset, clustering ethnic minorities. Our algorithms will then be trained on a very large dataset (representing the majority) while leaving out one or more ethnic minorities. We then apply Artificial Intelligence to the left-out minority. In a final step, we are interested in applying the established algorithm to local datasets.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-genotype-first-approach-in-medical-genomics-understanding-the-phenotypic-spectrum-of-monogenic-disorders

A genotype first- approach in medical genomics: understanding the phenotypic spectrum of monogenic disorders

Last updated:
ID:
551010
Start date:
4 February 2025
Project status:
Current
Principal investigator:
Dr Julie Rutten
Lead institution:
Leiden University Medical Centre, Netherlands

Research question
What is the frequency and phenotypic spectrum associated with deleterious variants in genes that are known to cause monogenic disorders in UK Biobank, and what are the genetic and environmental factors that influence disease expression?

We aim to address this question for the genetic disorders that are the focus of translational and fundamental research and patient care in the Leiden University Medical Center (LUMC) in the Netherlands. These LUMC-expertise monogenic disorders include: hereditary stroke and dementia syndromes, chromatinopathies, hereditary cancer, muscle disorders and DNA-repair disorders.

Objectives
To utilize linked genetic and phenotypic information in UK Biobank in order to:
1) determine the frequency of deleterious variants in genes associated with LUMC-expertise monogenic disorders
2) determine the phenotypic spectrum associated with these deleterious variants
3) identify genetic and environmental modifiers that play a role in disease expression

Data from UK Biobank will be combined with data from LUMC in-house patient registries, and with disease-relevant external databases. In this way, the full phenotypic spectrum is included, thereby increasing the power for the detection of modifying factors.

Scientific rationale
With the increasing use of whole exome and whole genome sequencing in routine clinical care, milder phenotypes associated with variants in genes known to cause severe monogenic disorders are increasingly recognised and identified. This creates challenges for patient and family counselling, but also opportunities for understanding disease pathomechanisms and identifying modifiers. An example is the research on NOTCH3 variants performed by the LUMC under UK Biobank application no. 74162, which has resulted in improved disease prediction in the clinic through the identification of a strong genotype-phenotype correlation (Rutten et al. Neurology 2020; Hack et al. Brain 2023).


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-genotypic-phenotypic-data-analysis-and-visualization-resource-to-study-normal-brain-aging

A genotypic-phenotypic data analysis and visualization resource to study normal brain aging

Last updated:
ID:
81764
Start date:
9 March 2022
Project status:
Current
Principal investigator:
Dr Kelsey Martin
Lead institution:
The Simons Foundation, Inc., United States of America

Even in the absence of neurodegenerative disorders like dementia or Alzheimer’s disorder, aging is associated with changes in the brain as well as deficits in cognitive ability. However, brain aging does not occur at the same pace or in the same way for all individuals. Some may age “optimally” with little to no changes in memory or cognition, while others may experience severe forms of impairment. This project aims to identify factors that are most important for maintaining brain health through late adulthood. We will study individual differences in genetics, lifestyle, and environment in combination with measures of brain shape, size and function. By characterizing which factors are most important for promoting longevity, this work will help craft public health policy and identify specific biological targets for medical research and intervention.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-genotyping-and-machine-learning-approach-towards-personalised-nutrition

A genotyping and machine learning approach towards personalised nutrition

Last updated:
ID:
55079
Start date:
12 June 2020
Project status:
Current
Principal investigator:
Mr Corentin Molitor
Lead institution:
Cranfield University, Great Britain

The goals of this project are to develop a list of DNA mutations linked to diabetes and obesity, which will be used to develop a personalised nutrition platform. Indeed, with the list of mutations, we can assess the potential risk a certain person has to develop these particular diseases. This risk scoring will be combined with clinical data as part of clinical studies to assess the effect of diet personalisation on young people with diabetes and obesity. The UK biobank data will be used to refine the list of DNA mutations and to develop the risk score models prior to the clinical studies.

The expected impact from this project is to motivate healthy dietary choices. This will be done by providing personalised nutritional advice, based on each person own information (DNA, biomarkers, clinical data!). Recent studies are showing that two persons can have a very different response to the same diet. This could explain why public health campaigns have not been effective in halting the rise in nutrition-related non-communicable diseases (NCDs) around the world. Notably because they rely on the “one size fits all” approach, which fails to take into account how each person will respond to a given diet. We expect personalised nutrition to provide a solution to this issue.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-graphical-interpretation-of-patient-data-in-the-uk-biobank

A Graphical Interpretation of Patient Data in the UK Biobank

Last updated:
ID:
80149
Start date:
9 March 2022
Project status:
Closed
Principal investigator:
Dr Tom Kai Him Leung
Lead institution:
Scinapsis Analytics Inc. dba BenchSci, Canada

Aim: We aim to add real-world evidence to our existing database, expanding relationships between different biological entities and, where applicable, assign a value to these relationships, which would inform us as to whether or not the relationship predicts a disease.

Scientific Rationale: Biological databases, generally, are created in isolation of one another by individual research groups. Consequently, Biological databases do not include relevant interactions with other biological databases. We extract information from separate databases, standardize it, creating one interconnected database. By linking related entries from these databases together, we have identified relationships that would otherwise not be captured if the databases were used independently, creating a web of related biological concepts. UK Biobank is an important data source of numerical experimental information on phenotypes, biomarkers, and diseases, which would improve the quality of the information provided by the relationships in our database.

Public Health Impact: The addition of real-world evidence to our existing database would provide important information on protein expression and its relationship to normal vs. disease states not available in traditional biological databases. This information will help pharma companies better understand the biology and molecular mechanisms for their target of interest. Further, our internal report showed that as much as 80% of preclinical experiments failed because of poor experiment design; the new network that we aim to develop will play a direct role in helping scientists execute better experiments.

Project Duration: The project duration will be 36 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-i-based-stroke-risk-factor-classification-and-treatment-abstract-study

A.I. Based STroke Risk fActor Classification and Treatment (ABSTRACT) study

Last updated:
ID:
783672
Start date:
4 November 2025
Project status:
Current
Principal investigator:
Dr William Heseltine-Carp
Lead institution:
University of Plymouth, Great Britain

Stroke is a leading cause of death and disability in the UK. Much of stroke management revolves around addressing risk factors with medications and lifestyle modification. However, 30% of those who suffer stroke have no known risk factors. Hence, there is need to better identify individuals who are at high risk of stroke, and particularly those where the benefit of treatment outweighs the risk.
ABSTRACT is a three phase study that looks to address this issue by (1) using artificial intelligence (AI) to predict stroke risk from routine hospital data, (2) to validate this model on external datasets, and (3) validate the ability to improve outcome by guiding clinical decision making.
Phase 1 has shown promising results, with Xgboost achieving an AUC of 94% when using routine blood test data, medical history and CT/MRI head imaging data to predict future stroke risk from a cohort of 9155 stroke cases and 109,875 controls in Southwest England. The model also identified several novel risk factors for stroke, such as liver function tests and C-reactive protein.

We now look to commence phase 2 of our project and validate these findings on an external dataset. At this time we ask the following research questions:
1. Can AI accurately predict stroke risk from routine hospital data?
2. How well does our model generalise to UK Biobank participants?
3. Does model performance vary across demographic and clinical subgroups?

Based on these questions, our research aims and objectives are therefore as follows:
1. To perform external validation of our stroke risk model using UKbiobank data.
2. To validate the novel risk factors for stroke identified by our models in phase 1.
3. To Assess model performance and calibration across different demographic subgroups

Following the results of this external validation we will then look to commence phase 3 of our project and assess the effectiveness of models in guiding clinical decision making and stroke risk management.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-large-scale-cohort-study-of-kidney-diseases-based-on-the-uk-biobank-database

A Large-Scale Cohort Study of Kidney Diseases Based on the UK Biobank Database

Last updated:
ID:
797261
Start date:
26 June 2025
Project status:
Current
Principal investigator:
Professor YongPing Lu
Lead institution:
The Seventh Affiliated Hospital of Sun Yat sen University, China

1. Integration of Multi-Omics Data and Exploration of Kidney Disease Mechanisms:
A comprehensive multi-omics analysis leveraging brain MRI, carotid ultrasound, and proteomic, genomic, and metabolomic profiles to identify risk factors and predictive markers for kidney disease and cardio-cerebrovascular events, with an emphasis on the pathophysiology of cardiovascular-kidney-metabolic (CKM) syndrome.
2. Investigating Genetic and Environmental Risk Factors for Kidney Diseases:
Combine UK Biobank genomic data (whole-genome sequencing, genotyping) with environmental exposure data (air pollution, lifestyle factors, etc.) to analyze how gene-environment interactions impact the development of kidney diseases, such as chronic kidney disease and acute kidney injury.
3. Develop an Early Warning Model for Kidney Disease:
Utilize longitudinal data from UK Biobank (including biochemical markers and imaging data) along with electronic health records (linked via the NHS) to build risk prediction models based on machine learning or deep learning techniques.
4. Evaluate the Long-Term Effects of Medications and Treatment Strategies:
Analyze medication records and follow-up data from UK Biobank to assess the impact of renin-angiotensin system inhibitors (ACEI/ARB) and novel targeted therapies (e.g., rituximab) on the progression of kidney diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-large-scale-multi-omics-cohort-study-of-hemorrhagic-stroke-integrating-neuroimaging-genetics-and-vascular-risk-factors-via-the-uk-biobank

A Large-Scale Multi-Omics Cohort Study of Hemorrhagic Stroke: Integrating Neuroimaging, Genetics, and Vascular Risk Factors via the UK Biobank

Last updated:
ID:
952957
Start date:
6 August 2025
Project status:
Current
Principal investigator:
Dr Feng Lu
Lead institution:
Fujian Medical University, China

Through comprehensive multi-dimensional data analysis, we will deeply explore the pathogenesis of hemorrhagic stroke and develop an early warning model:
1. Integrate genomics (whole genome sequencing, exome), proteomics (plasma protein measurement), metabolomics (serum metabolites) and neuroimaging data (brain MRI/CT) to reveal the molecular network of hemorrhagic stroke. This will help identify new biomarkers and clarify the interactions between genetic susceptibility, vascular inflammation pathways, coagulation dysfunction, and blood-brain barrier integrity.
2. Combining genomic data (whole genome sequencing, genotyping) and vascular risk factors (hypertension, lifestyle, dietary patterns) to construct a polygenic risk score (PRS). PRS can quantify the combined effects of genetic variants (such as APOE, COL4A1 gene mutations) and environmental factors on hemorrhagic stroke risk and help identify high-risk populations for preventive interventions.
3. Using brain imaging data (MRI quantification of cerebral small vessel disease, microbleeds detection) and longitudinal health records (blood pressure monitoring, coagulation parameters), develop a deep learning-based system to automatically identify imaging biomarkers of hemorrhagic stroke risk. Combined with clinical indicators, a dynamic risk prediction model is created to detect potential hemorrhagic events early for timely intervention and improved patient outcomes.
In general, I will comprehensively analyze the pathogenesis of hemorrhagic stroke, identify new biomarkers, and develop early warning models through multi-omics mechanism analysis, gene-environment interaction research, and neuroimaging phenotype analysis to provide a scientific basis for clinical prevention and treatment.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-large-scale-multi-omics-cohort-study-of-pulmonary-fibrosis-integrating-genomics-imaging-and-environmental-exposures-via-the-uk-biobank

A Large-Scale Multi-Omics Cohort Study of Pulmonary Fibrosis: Integrating Genomics, Imaging, and Environmental Exposures via the UK Biobank

Last updated:
ID:
855274
Start date:
23 July 2025
Project status:
Current
Principal investigator:
Dr Andong He
Lead institution:
The Affiliated Lihuili Hospital of Ningbo University, China

Through multi-dimensional data analysis, we will deeply explore the pathogenesis of pulmonary fibrosis and develop an early warning model:
1. Integrate genomics (whole genome sequencing, exome), proteomics (plasma protein measurement), metabolomics (serum metabolites) and imaging data (chest CT/MRI) to reveal the molecular network of pulmonary fibrosis. This will help identify new biomarkers and clarify the interactions between genetic susceptibility, inflammatory pathways and metabolic abnormalities.
2. Combining genomic data (whole genome sequencing, genotyping) and environmental exposure data (air pollution, lifestyle, etc.) to construct a polygenic risk score (PRS). PRS can quantify the combined effects of genetic and environmental factors (such as MUC5B gene mutation and smoking) on !!the risk of pulmonary fibrosis and help identify high-risk groups.
3. Using chest imaging data (CT quantification of interstitial lung changes) and longitudinal health records (pulmonary function, hospitalization data), develop a deep learning-based system to automatically identify imaging biomarkers of pulmonary fibrosis. Combined with clinical indicators, a dynamic risk prediction model is created to detect pulmonary fibrosis early so that timely intervention can improve patient prognosis.
In general, I will comprehensively analyze the pathogenesis of pulmonary fibrosis, identify new biomarkers, and develop early warning models through multi-omics mechanism analysis, gene-environment interaction research, and imaging phenotype analysis to provide a scientific basis for clinical diagnosis and treatment.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-large-scale-study-in-uk-biobank-reveals-the-genetic-metabolic-and-clinical-panorama-of-metabolic-dysfunction-associated-fatty-liver-disease

A large – scale study in UK Biobank reveals the genetic, metabolic and clinical panorama of metabolic – dysfunction – associated fatty liver disease

Last updated:
ID:
985626
Start date:
16 October 2025
Project status:
Current
Principal investigator:
Miss Hong Ren
Lead institution:
Zhongshan Hospital Affiliated to Fudan University, China

Research questions: Metabolic-associated steatosis liver disease (MASLD), as the new name for non – alcoholic fatty liver disease (NAFLD), still requires a comprehensive elucidation of its complex pathophysiological mechanisms, diverse clinical phenotypes, and the synergistic effects with metabolic diseases such as cardiovascular diseases and type 2 diabetes. In large-scale population cohorts, how to accurately identify susceptible populations, make early diagnoses, and effectively intervene to block disease progression remains a key scientific issue that urgently needs to be addressed. How does the genetic basis of MASLD affect its occurrence and development? What is the interaction mechanism between different metabolic disorders (such as obesity, diabetes) and MASLD? Do significant differences exist in the clinical characteristics and prognosis of MASLD patients under different BMI classifications (e.g., lean vs. overweight/obese)? What are the effectiveness and limitations of existing diagnostic markers and
treatment strategies in the real world?
Research objectives: This study aims to utilize the large-scale, multi-dimensional, and prospective population cohort data of the UK Biobank to deeply analyze the pathological mechanisms, clinical characteristics of Metabolic-associated steatosis liver disease (MASLD), and its association with cardiometabolic diseases. On this basis, potential diagnostic and treatment strategies will be explored.
Scientific rational: MAFLD involves complex pathological mechanisms centered on hepatic steatosis, linked to insulin resistance, inflammation/oxidative stress, lipid metabolism disorders, and gut-liver axis dysfunction. Clinically, it ranges from asymptomatic to advanced liver disease, associates with metabolic diseases, includes lean cases (BMI<25), and increases cardiovascular risk and sarcopenia likelihood.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-life-course-approach-investigating-genetic-and-lifestyle-associations-contributing-to-cardiovascular-disease-risk

A life course approach investigating genetic and lifestyle associations contributing to cardiovascular disease risk

Last updated:
ID:
82209
Start date:
30 March 2022
Project status:
Current
Principal investigator:
Professor Bernard Keavney
Lead institution:
University of Manchester, Great Britain

We will use multiple UK Biobank resources to better understand the relationship between both genetic and environmental factors with cardiovascular diseases including congenital heart disease. Over the course of the project we aim to discover novel associations influencing disease risk, the reasons why some other diseases occur together with cardiovascular disease, and discover factors that influence the severity of disease over time. This could ultimately impact future healthcare as it may enable earlier diagnosis and treatment in families affected by these conditions, improving the development of personalised care pathways.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-lifecourse-approach-to-womens-health-utilising-pregnancy-as-an-opportunity-to-prevent-ill-health

A lifecourse approach to women’s health: utilising pregnancy as an opportunity to prevent ill health

Last updated:
ID:
708764
Start date:
4 August 2025
Project status:
Current
Principal investigator:
Dr Julia Zollner
Lead institution:
University College London, Great Britain

Scientific rational for research
Evidence links several pregnancy-related conditions to long-term health risks, including gestational hypertension and cardiovascular disease, gestational diabetes and metabolic disorders, and perinatal mental health issues with ongoing psychological needs. Despite these well-established associations, there is limited understanding of how pregnancy complications translate into long-term health trajectories and when intervention is most effective.
More precise models for risk prediction, incorporating a range of health and ‘omic factors, could enable earlier identification of high-risk individuals. By leveraging pregnancy as a predictor of long-term health, there is an opportunity to improve preventative strategies, personalise care, and enhance health outcomes across the life course.

Research question
Can the utilisation of ‘omics data incorporated alongside traditional clinical risk factors predict future ill health after pregnancy complications?

Objectives
1. Assess long-term disease risk following pregnancy complications
– Evaluate the progression from gestational diabetes to type 2 diabetes
– Investigate how hypertensive disorders of pregnancy (e.g., preeclampsia, gestational hypertension) influence future cardiovascular disease risk
2. Examine the impact of pre-existing conditions on pregnancy-related disease progression
– Determine how pre-existing cardiometabolic conditions (e.g., obesity, chronic hypertension, type 1 or 2 diabetes) during pregnancy influence long-term health trajectories
3. Utilise longitudinal clinical and biochemical data to define disease trajectories
– Establish patterns of onset and progression of diabetes, cardiovascular disease, and related metabolic conditions
4. Leverage multi-omics data to explore genetic and molecular determinants of disease
– Use whole genome sequencing, exome sequencing, and proteomic data to investigate genetic drivers of metabolic and cardiovascular disease


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-light-weight-interpretable-deep-learning-framework-for-cardiomyopathy-diagnosis-based-on-multimodal-data

A light-weight interpretable deep learning framework for cardiomyopathy diagnosis based on multimodal data

Last updated:
ID:
301899
Start date:
14 May 2025
Project status:
Current
Principal investigator:
Dr Peifang Zhang
Lead institution:
Biomind Technology Co. Ltd, China

Cardiomyopathy is a disease of the heart muscle. It causes the heart to have difficulties to pump blood to the whole body, which can lead to symptoms of heart failure. Cardiomyopathy also can lead to sudden cardiac death. In China, it is estimated in 2023 that there are about 1,000,000 patients suffering hypertrophic cardiomyopathy and about 120,000 patients suffering dilated cardiomyopathy. According to “2024 heart disease and stroke statistics: a report of US and global data from the American Heart Association”, 410,000 deaths were estimated for cardiomyopathy and myocarditis based on 204 countries and territories in 2021.
Since genetic testing is costly and has a long report generation period, the phenotypic cardiac MRI (CMR) imaging is usually considered as the necessary examination utility for cardiomyopathy diagnosis, as it can be used to measure the cardiac structure and function, evaluate the severity of cardiomyopathy. However, the analysis of cardiac MRI imaging is a heavy workload to physicians as it requires a lot of time to contour heart anatomy and requires long time training to improve their accuracy. For the similar reasons, only two commercial software tools are available over the world for CMR analysis for a long time.
Beyond the CMR imaging, clinical data such as baseline and lab test variables are also important for cardiomyopathy diagnosis. So given the amazing progress in deep learning techniques, we aim to apply deep learning on multi-modal medical data to diagnose various forms of cardiomyopathies.
However, in the medical application scenario, large deep learning models are inherently black boxes, and hard to gain the trust of medical professionals. We break the large black-box into 3 pieces and have medical knowledge embedded in the framework, so that we not only take advantage of the deep learning advances, but also provide an opportunity for medical professionals to trust AI.
Furthermore, our knowledge oriented deep learning framework achieved high diagnosis accuracy to benefit cardiomyopathy patients and could run in portable or small computing devices.
Through our research project, the public attention may be directed to other possibilities than the overwhelming large AI models, especially in the medical AI region. Our knowledge oriented approach perfectly avoid the illusion pitfalls that most large AI models currently face.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-longitudinal-study-of-chronic-disease-onset-progression-and-overdiagnosis

A Longitudinal Study of Chronic Disease Onset, Progression, and Overdiagnosis

Last updated:
ID:
506132
Start date:
27 November 2024
Project status:
Current
Principal investigator:
Dr Yihui Du
Lead institution:
Hangzhou Normal University, China

This research dives into the development and progression of chronic diseases, including, but not limited to, cardiovascular disease, respiratory disease, metabolic disease, and cancer. It aims to 1) track how individuals transition from healthy states to single chronic disease, then multimorbidity, and ultimately death. 2) identify distinct patterns in this progression, considering which diseases co-occur and the order of their appearance. 3) investigate the causal relationships between lifestyle factors, genetic predispositions, and the development and progression of chronic diseases. Identify potential mediators and modifiers that influence these pathways. 4) assess the prevalence and impact of overdiagnosis in chronic diseases, including the identification of factors contributing to overdiagnosis.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-longitudinal-study-on-clonal-hematopoiesis-of-indeterminate-potential-chip-and-its-impact-on-diabetic-micro-vascular-complications

A longitudinal study on clonal hematopoiesis of indeterminate potential (CHIP) and its impact on diabetic micro-vascular complications

Last updated:
ID:
786986
Start date:
23 July 2025
Project status:
Current
Principal investigator:
Dr Byung-Wan Lee
Lead institution:
Yonsei University, Korea (South)

CHIP is an age-related phenomenon characterized by somatic mutations in hematopoietic stem cells. These mutations lead to the clonal expansion of mutant blood cells without overt hematologic malignancies. While CHIP is primarily associated with an increased risk of atherosclerotic cardiovascular diseases (ASCVD), emerging evidence suggests a potential role in metabolic disorders, including type 2 diabetes mellitus (T2DM) and related complications. Prior studies on CHIP in relation to T2DM have demonstrated an increased prevalence and incidence of T2DM. Although there have been individual cohort studies on the risk of CHIP on developing diabetic complications, evidence from large cohort studies is lacking. The increased risk and mortality pertaining to ASCVD, we hypothesized microvascular complications, especially diabetic kidney disease and its associated phenotype of albuminuria and proteinuria, to be influenced by the aberrant immunologic response in CHIP carriers with diabetes.
We thus aim to analyze individuals with whole-exome sequencing data to compare the incidence of diabetes-related complications (e.g., nephropathy, retinopathy, neuropathy) between CHIP carrier T2DM individuals with non-CHIP carriers T2DM individuals, and baseline non-diabetics with or without CHIP who developed T2DM during follow-up


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-machine-learning-based-10-year-risk-prediction-model-for-psoriasis-using-metabolomics

A machine learning-based 10-year risk prediction model for psoriasis using metabolomics

Last updated:
ID:
952292
Start date:
11 September 2025
Project status:
Current
Principal investigator:
Ms Yi Wei
Lead institution:
Zhujiang Hospital of the Southern Medical University, China

1.Research question
The diagnosis of psoriasis faces multiple challenges. Its skin lesions are highly similar to seborrheic dermatitis and eczema, easily causing confusion and misdiagnosis. Early symptoms are mild and subtle, often overlooked and delayed. Lacking specific biological indicators, diagnosis relies on clinical manifestations and historical experience. With significant symptom differences among subtypes and dynamic changes in lesions during progression, difficulties increase, demanding specific biomarkers to enhance diagnostic efficacy.
2.Objectives of the study
To construct a 10-year psoriasis risk prediction model based on metabolomics for psoriasis risk stratification.2.To explore the predictive value of key predictors for the risk of psoriasis.
3.Scientific basis
Plasma metabolites can directly reflect the physiological and pathological status of the organism, with dynamic sensitivity and easy detectability, providing specific molecular markers for diseases; meanwhile, machine learning excels at mining potential associations from massive high-dimensional metabolomics data, overcoming the limitations of manual analysis to improve the efficiency of marker screening and diagnostic model construction.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-machine-learning-based-prs-model-predicting-risk-of-developing-diabetic-nephropathy-and-hypertension-in-diabetes-patients

A machine-learning based PRS model predicting risk of developing diabetic nephropathy and hypertension in diabetes patients

Last updated:
ID:
95961
Start date:
15 June 2023
Project status:
Current
Principal investigator:
Miss Dongyan Yang
Lead institution:
Northwest University, China

The diabetic and hypertensive-related nephropathy is a serious life-threatening condition. Catching the condition early and taking actions in advance can help slow down or even stop the progression of these diseases. Diabetic DN and hypertension are affected by many genetic changes, and non-genetic features frequently mentioned as social and environmental factors. A polygenic risk score is a score that can estimate a person’s risk for developing a disease based on their genetics or DNA. This is often displayed in a way that tells how at risk a person is for a specific condition. The proposed study is to investigate these changes and factors to understand the role that genetic and environmental factors play in developing diabetic DN and hypertension across different populations. The expected value of this study is to produce a novel scoring system to predict the risk of developing diabetic DN and hypertension in diabetes patients, based on the total number of changes and other information that is related to this condition. Our goal is to provide an accurate risk assessment tool that can help diabetes patients to manage their personal risk for developing diabetic DN and hypertension. The proposed duration of this study is 2 or 3 years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-machine-learning-based-screening-system-for-retinal-and-systemic-diseases

A Machine Learning based screening system for Retinal and Systemic Diseases

Last updated:
ID:
44226
Start date:
3 January 2019
Project status:
Current
Principal investigator:
Dr Alauddin Bhuiyan
Lead institution:
iHealthScreen Inc, United States of America

Latest advances in machine learning, specifically the deep learning technique have shown tremendous potential in medical applications for diagnosing and detecting diseases at early stage based on imaging data. In this research project, we aim to make use of these advances to automatically identify people at risk of certain eye and systemic diseases such as Diabetic Retinopathy, Age-related Macular Degeneration, Stroke and Cardiovascular diseases at early stage and help prevention. For example, the number of Americans with diabetic retinopathy is expected to rise from 7.7 million to 14.6 million between 2010 and 2050. It is estimated that nearly 80% of these cases can be preventable. Similarly, By 2050, the estimated number of people with AMD is expected to more than double from 2.07 million to 5.44 million. Early detection is key to prevent these diseases and reducing the unnecessary overhead of medical bills, and save sight and lives. The project will make use of the vast imaging and demographic data that UK Biobank has, to build such systems to predict the onset of these diseases. We estimate that it will take up to two years to complete the research project.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-machine-learning-platform-applied-to-multi-modal-data-to-uncover-risk-factors-for-chronic-pain-conditions-to-direct-mechanistic-studies

A machine learning platform applied to multi-modal data to uncover risk factors for chronic pain conditions to direct mechanistic studies.

Last updated:
ID:
93141
Start date:
4 April 2023
Project status:
Current
Principal investigator:
Mr Michael Robert Allwright
Lead institution:
University of Sydney, Australia

Our project uses modern machine learning techniques to predict whether people will develop certain diseases, specifically focussing on chronic pain, such as diabetic polyneuropathy, Chronic Regional Pain Syndrome (CRPS) and chronic lower back pain. We aim to use this platform to help understand the reasons why people develop those conditions and to inform further research into candidate mechanisms through which chronic pain occurs. By using the large sample size available through the UK Biobank (UKB, n > 500,000), we will determine for example whether certain blood biomarkers are co-incident with chronic pain onset, which genes are most highly associated with chronic pain, and what is the intersection between genetics, lifestyle, biomarkers and brain structure in predicting chronic pain conditions.

This research supports a movement towards more personalised medicine by helping researchers develop a deeper and more bespoke understanding of which individuals are at higher risk of chronic pain based on clustering populations according to both genetics and phenotype. Through this research we hope to cast light upon how different aspects of pain affect different people, based on their genetics, biometrics, brain scans and many other features.

The UKB is unique in the sense that it contains multi-modal data for a range of variables across a large number of individuals. This means that for the first time it is feasible to build models that look across thousands of variables without pre-supposing any particular association, to uncover novel risk factors for a disease of interest. As a result of this it will be possible to firstly validate the existing factors responsible for a disease, but in addition to both identify new factors and uncover more complex connections between different factors, for example the importance of certain inflammatory biomarkers in predicting chronic pain.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-mendelian-randomisation-study-to-assess-the-interaction-between-adcy9-rs1967309-hdl-diet-and-atherosclerosis-measured-by-carotid-intima-media-thickness-cimt

A Mendelian Randomisation Study to Assess the Interaction between ADCY9 rs1967309, HDL, Diet and Atherosclerosis Measured by Carotid Intima Media Thickness (CIMT).

Last updated:
ID:
25602
Start date:
14 May 2018
Project status:
Closed
Principal investigator:
Dr Patrick Gladding
Lead institution:
Waitemata District Health Board, New Zealand

Raising HDL, using medication, has not been shown to improve cardiovascular outcomes, despite epidemiological studies showing that high HDL is beneficial. A recent genome-wide-association study has shown that the HDL raising drug, dalcetrapib, reduces cardiovascular events and atheroma in patients with a particular genotype. This effect may be either gene-drug or gene-HDL related. We propose a mendelian randomisation study to evaluate the relationship between de novo HDL levels and the ADCY9 rs1967309 and its influence on carotid artery intima media thickness (CIMT), as a marker of atherosclerosis. UK Biobank’s stated purpose is to support health-related research that is in the public’s interest. Our study will have wide relevance to population health given the high prevalence of atherosclerosis, and morbidity/mortality associated with the disease. If a relationship is shown to exist between HDL and the ADCY9 gene, which is independent of dalcetrapib, then this drug-gene association will be more widely relevant to the population, even in the absence of this drug. Both the drug and the gene test are patented. No biological samples in the UK Biobank will be used in this study. Subjects will be identified by their HDL levels. Subjects who have undergone CIMT testing will be stratified by their HDL levels. Subjects in the lowest and highest HDL quartile will be evaluated to test the relationship between CIMT and the ADCY9 gene. This project can be completed with only electronic data and will not require consumption of biological samples. The CIMT subset of the full cohort will be used. This study will only be possible if either the ADCY9 rs1967309 SNP has been tested on the UK Biobank array or can be imputed from the existing data.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-mendelian-randomization-analysis-of-healthy-habits-and-outcomes-using-international-genomic-cohorts

A Mendelian Randomization Analysis of Healthy Habits and Outcomes Using International Genomic Cohorts.

Last updated:
ID:
148863
Start date:
7 March 2024
Project status:
Current
Principal investigator:
Dr Keita Hirano
Lead institution:
Kyoto University, Japan

Aims:
We aim to investigate and clarify the direct links between health habits and their effects on cardiovascular and renal risks. Using the Mendelian randomization method, we hope to gain a clearer understanding of these connections, which traditional studies might miss due to possible biases.

Scientific Rationale:
It’s often challenging in contemporary health research to clearly determine the direct links between habits and health outcomes. Our approach is different because we use Mendelian randomization, a method that uses genetic differences as tools, to identify direct causes without the usual interfering factors. We plan to use the detailed data from the UK Biobank, especially the GWAS data, to find specific genetic markers linked to health habits.

With this genetic information and a wide range of data on diet, health, and background factors, we can create studies that mimic randomized controlled trials. This will help us estimate the direct effects of health habits on heart and kidney health outcomes. The broad and detailed nature of the UK Biobank data means our analysis can consider a wide range of diets, backgrounds, and health outcomes.

Project Duration:
We expect the project to last three years. The first year will be about gathering data and an initial analysis. The next year will focus on the Mendelian randomization and more detailed studies. The last year will be for interpreting the data, confirming our results, and sharing our findings.

Public Health Impact:
The results of our study could be very important for public health. With growing concerns about heart and kidney diseases, it’s essential to understand the direct links between our daily habits and these health problems. Using the detailed UK Biobank data, we aim to give more specific and evidence-based health advice.

By creating health improvement programs and personalized recommendations based on our findings, we hope to improve public health and well-being. This study could also guide future health research by showing the importance of using wide-ranging and detailed data for creating evidence-based health advice.

In Conclusion:
For our study to be successful, we need to access various types of information from the UK Biobank. This request highlights the importance of having a wide range of data. It shows that a thorough approach, based on detailed and varied data, can produce strong health research that can be applied in many ways.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-meta-analytic-genome-wide-association-study-for-vitamin-d-levels

A meta-analytic genome-wide association study for Vitamin D Levels

Last updated:
ID:
16009
Start date:
1 February 2016
Project status:
Closed
Principal investigator:
Professor Nicholas Timpson
Lead institution:
University of Bristol, Great Britain

We propose to run a large scale analysis of genome-wide association studies (GWAS) to identify novel common, low-frequency and rare genetic variants associated with vitamin D (specifically 25-hydroxyvitamin D (25OHD)). We are aiming to expand the understanding of how genetic variation contributes to levels of vitamin D which is known to be associated with a host of different disorders and also with biological pathways important for diseases such as cancer. This understanding will not only help us to understand what determines levels, but also apply this knowledge to assess the impact of vitamin D levels on disease. Vitamin D insufficiency affects up to half of otherwise healthy adults with detrimental impacts on public health. Approximately 50% of the variability in 25OHD levels is attributable to genetic factors. Although genetic factors are speculated to contribute substantially to this variability, the identified to date four common variants explain little of the heritability. The objective of the present study is to detect additional common, low-frequency and rare variants in novel genetic loci of large effect, associated with 25OHD levels, enabling the identification of novel pathways implicated in 25OHD metabolism and groups of individuals at risk for 25OHD insufficiency. We have collected 12 participating studies, with approximately 30,000 individuals with genome-wide genotypes and 25OHD levels. All cohorts have assessed the effect of genetic variants on 25OHD levels. We have imputed each cohort to the UK10K/1000Genomes reference panel which enables more accurate estimation of low frequency and rare genotypes. While we have identified preliminarily interesting findings, we aim to include the UKBiobank data to replicate our findings and identify potential novel loci. To do so, we will undertake a fixed-effects meta-analysis of all cohorts of the effect of each genetic variant on 25OHD levels. For these analyses we request access to genome-wide genotyping data, serum levels of 25OHD, and the following covariates: sex, age, body mass index, date of vitamin D measurement and reported vitamin D intake through food and supplemental sources. Specifically we request the above data for the entire UKBiobank cohort as and when available. We would like to use and extend (with the full release when available) imputed GWAS data generated as part of UKBiobank project 8786. This will greatly speed up our project and prevent replication of effort and minimise the amount of storage we use for this project.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-meta-investigation-of-combinatorial-mutation-signatures-in-broad-disease-categories

A meta-investigation of combinatorial mutation signatures in broad disease categories

Last updated:
ID:
44288
Start date:
20 November 2018
Project status:
Current
Principal investigator:
Dr Sayoni Das
Lead institution:
PrecisionLife Ltd, Great Britain

The proposed study aims to address three research questions:

* Are there genetic defect patterns common to broad categories of disease such as all cancers, all psychiatric disorders, or all musculoskeletal disorders?
* Conversely, are there genetic patterns that help protect people against broad categories of disease, such as cancer or cardiovascular disease?
* Are there genotypic variant signatures allowing stratification of patients that could inform the risk of developing a disorder and likelihood of drug therapy response?

The proposed research would improve our understanding of the genomic basis of disease formation (or disease prevention). We hope to identify individual genetic defects or combinatorial defect clusters that are commonly associated with broad categories of disease, that is, found in significantly higher numbers of patients compared to healthy controls. Similarly, we hope to identify protective signatures that are found in many more healthy controls compared to afflicted individuals.

In addition, we hope to develop new improved ways of identifying patients at risk of developing a disease or its complications, and enable patients to be treated with a drug therapy regimen that is tailored to their individual needs. Successful results would help future researchers identify means to increase human longevity and wellness by manipulating genetic mechanisms involved in broad categories of disease. Through follow-on studies, researchers may identify new drugs that work across broad categories of disease. Such drugs with broad applicability may cost less to develop, test, and bring to market, thus helping everyone afflicted with those diseases. Tailoring therapies to patients’ individual needs may significantly reduce the burden on the healthcare system through reduced side effects due to drug interaction or lack of therapy response, leading to hospital admissions.

The project duration is 24 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-metabolic-profile-of-muscle-function-in-middle-aged-and-older-adults

A Metabolic Profile of Muscle Function in Middle-aged and Older Adults

Last updated:
ID:
102947
Start date:
8 May 2025
Project status:
Current
Principal investigator:
Dr Keith Avin
Lead institution:
Indiana University Purdue University Indianapolis, United States of America

Aims- Aim 1. To develop a metabolite profile of muscle function in a cross-sectional analysis of middle and older aged adults. Aim 2: To validate metabolite profiles of muscle function that are predictive of muscle function change over time.

Scientific rationale- Sarcopenia is a condition that may occur with aging that results in muscle weakness and loss. Weak and smaller muscles are linked to an increased risk in disability, chronic disease and mortality. Sarcopenia is treated with physical activity and exercise. The focus only on activity suggests that each person can control whether their muscles get smaller and weaker. We believe that muscle strength and size are not only impacted my activity, but metabolism as well. Our goal is to identify metabolic markers from people who appear healthy but demonstrate either poor, average, or good muscle function. If metabolic disturbances can be identified during middle age, it can then help prevent changes with aging. Muscle function is difficult to fully characterize given 600 muscles uniquely responding to lifestyle choices, aging, disease, pharmacology and inter-organ and tissue crosstalk such as with liver, bone, and heart. Therefore, we are utilizing a systemic approach by analyzing blood metabolites of individuals with above average, average, and below average muscle function between age categories of 40-59 and >60 years old. The metabolic profile will identify those who are at risk and should be intervened at an earlier timepoint in future studies. Collectively, this data will establish multiple proposals that will identify those who are at long-term risk for muscle dysfunction and to personalize exercise prescription in future studies.

Expected duration of project- 2 years

Public health impact of the work- Identifying metabolic contributions to poor muscle health can provide direction on how to best manage physical activity and other factors such as nutrition and nutraceuticals.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-metabolomics-foundation-model-for-cross-study-biological-age-estimation-and-disease-prediction

A Metabolomics Foundation Model for Cross-Study Biological Age Estimation and Disease Prediction

Last updated:
ID:
786652
Start date:
24 June 2025
Project status:
Current
Principal investigator:
Dr Yuanxu Gao
Lead institution:
Peking University, China

I. Research Questions
1. How can we develop a metabolomics-specific foundation model to capture the complex relationships between metabolites and aging/disease outcomes?
2. Can a foundation model effectively predict biological age and disease risk by integrating multimodal data, including metabolic profiles and environmental factors?
3. What are the key metabolic pathways driving population-specific aging patterns, particularly influenced by diet and environmental exposures?

II. Objectives
1. Develop a metabolomics-specific foundation model to harmonize heterogeneous data and capture complex aging patterns.
2. Fine-tune the model for biological age estimation and disease risk stratification using UK Biobank data.
3. Identify key metabolic pathways driving population-specific aging patterns using interpretability techniques.

III. Scientific Rationale
1. Data Heterogeneity: Existing metabolomics studies are hindered by data heterogeneity. A foundation model can harmonize diverse datasets, improving generalizability.
2. UK Biobank Data: The UK Biobank’s extensive longitudinal data provides a unique opportunity to develop a robust model for aging and disease prediction.
3. Clinical Relevance: Fine-tuning the model for biological age and disease risk will provide actionable insights for early detection and personalized interventions.

IV. Plan for Disseminating Our Findings
We will disseminate our findings through high-impact SCI publications and presentations at international conferences to engage the academic community. In addition, we will develop and release an open-source web tool based on our findings, enabling the public and researchers to explore and apply the model. Our goal is to make our results accessible through both academic and public channels, ensuring broad impact and contributing to the field.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-model-for-health-risk-prediction-based-on-inherited-dna-variation-and-clinical-data

A model for health risk prediction based on inherited DNA variation and clinical data

Last updated:
ID:
88907
Start date:
14 September 2023
Project status:
Current
Principal investigator:
Dr Mykyta Artomov
Lead institution:
Nationwide Childrens Hospital, United States of America

Aging is commonly associated with an increase in the risk of complex phenotypes onset, such as cardiovascular diseases, diabetes, cancer, etc. One can consider healthy aging or mortality as complex phenotypes shaped by susceptibilities to many underlying diseases and with significant contribution of environmental factors. In this context, the outcomes could be defined as age of major disease onset, age of critical health deterioration or age of death. We will use such interpretations to build a predictor for the individual risks of various aging outcomes. Specifically, we will start with most common diseases of aging – cardiovascular disease (and incidents) and cancer. Further, the mathematical design of the predictor will be investigated in other phenotypes – diabetes, autoimmune disorders, to understand specific features required for each disease type.

Main deliverables are: first, information about feature importance, such as, selection of data points that are most informative for health outcome. Second, average health endpoints for individuals with similar input parameters, found in training data. And finally, data-based individual longevity recommendations for managing parameters with the highest contribution to age/mortality prediction. This work is in line with the UK Biobank’s aim of enabling research to improve prevention, diagnosis and treatment of illness and the promotion of health throughout society.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-multi-level-approach-to-better-understand-the-association-between-physical-activity-and-sedentary-behaviour-and-cancer-risk

A multi-level approach to better understand the association between physical activity and sedentary behaviour, and cancer risk.

Last updated:
ID:
3173
Start date:
1 March 2017
Project status:
Closed
Principal investigator:
Dr Ruth Hunter
Lead institution:
Queen's University Belfast, Great Britain

Our aims are to investigate physical activity behaviour and sedentary behaviour, and cancer risk, including better estimations of the magnitude of risk by different cancer sites. Specifically, we aim:
1) To evaluate correlations between physical activity and sedentary behaviour data and relevant biomarkers related to physical activity/sedentary behaviour and cancer
2) To analyse the association between physical activity and sedentary behaviour data and relevant genetic variants identified within GWAS data and cancer risk.
3) To analyse factors related to the built environment, physical activity, sedentary behaviour, and cancer risk.
The aim of UK Biobank is to improve the prevention, diagnosis and treatment of serious and life-threatening illnesses, including cancer. The proposed research will elucidate the role of physical activity and sedentary behaviour in cancer prevention. The study will provide insight into biological mechanisms by which physical activity and sedentary behaviour may impact on different cancer sites, the role of individual modifiable and non-modifiable characteristics, and built environment factors in influencing the association between physical activity, sedentary behaviour, and cancer risk. Results will help inform the development of future physical activity/sedentary behaviour related interventions for cancer prevention. Linkage between UK Biobank and cancer registries provides information on cohort members who have been diagnosed with cancers. Using appropriate statistical techniques, we will compare physical activity and sedentary behaviour amongst cohort members who have developed cancer with those who have not developed these cancers, examining questionnaire and accelerometer (measure of physical activity/sedentary behaviour) data. We will also evaluate the associations with relevant biomarkers from the panel of blood/urinary markers and genetic analyses to be undertaken on all cohort members. The full cohort will be included in all analyses to be undertaken.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-multi-modal-ai-foundation-model-to-advance-precision-medicine

A Multi-Modal AI Foundation Model to Advance Precision Medicine

Last updated:
ID:
933830
Start date:
29 July 2025
Project status:
Current
Principal investigator:
Dr George Okafo
Lead institution:
Standard Model Biomedicine Inc, United States of America

AIM – Over the next three years, we will develop a versatile AI foundation model that integrates health records, medical images, and genomic data from the UK Biobank. Our goal is to set a new standard in predicting disease risk, forecasting outcomes, and uncovering biological drivers of both common conditions (like cancer) and rare disorders.
BACKGROUND & RATIONALE – Today, clinicians must sift through vast amounts of patient history, imaging scans, lab tests, and genetic information to make treatment decisions. As biomedical data grows, this process becomes more time-consuming and prone to oversight. AI offers the promise of identifying hidden patterns across diverse data types, but existing tools rarely handle records, images, and genomic sequences all at once-especially at the scale of hundreds of thousands of patients. By training our model on such a large, varied population, we expect it to perform accurately for individuals from many genetic and environmental backgrounds.
PUBLIC HEALTH IMPACT – The conditions represented in the UK Biobank affect millions worldwide. Our model will help predict who is at highest risk, estimate likely disease trajectories, and suggest underlying molecular causes. We will share our code and results openly, so other researchers can build on our work. Ultimately, this will lead to earlier testing and diagnosis, more personalized treatment plans, and new insights that drive the discovery of novel therapies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-multi-modal-approach-for-exploring-brain-reorganization-in-age-related-hearing-loss-and-its-relationship-to-cognitive-decline

A multi-modal approach for exploring brain reorganization in age-related hearing loss and its relationship to cognitive decline

Last updated:
ID:
65795
Start date:
26 February 2021
Project status:
Closed
Principal investigator:
Dr Maria J. S. Guerreiro
Lead institution:
Carl von Ossietzky University Oldenburg, Germany

Age-related hearing loss has been shown to be associated with greater cognitive impairment among older adults; however, the mechanisms underlying this relationship are not completely understood. This project aims to investigate the potential brain mechanisms and pathways linking age-related hearing loss and cognitive decline in older adults, by examining the effect of age-related hearing loss on different measures of brain organization and how these, in turn, relate to cognitive decline. This project will run for a 36-month period, and is expected to increase our understanding of the cognitive and neurobiological correlates of age-related hearing loss, while generating hypotheses for future interventional studies examining the potential benetfits of hearing interventions on brain organization and cognitive performance.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-multi-modal-deep-learning-strategy-to-predict-glaucoma-onset-and-progression

A multi-modal deep learning strategy to predict glaucoma onset and progression

Last updated:
ID:
129271
Start date:
28 February 2024
Project status:
Current
Principal investigator:
Dr Yi Zhou
Lead institution:
Xiangya Hospital of Central South University, China

1. Aims:
1.1. To develop a deep learning algorithm for glaucoma using patients’ clinical assessment, epidemiologic data, living environment record and imaging.
1.2. To explore the genetic and metabolomics factors associated with the mechanisms of glaucoma pathogenesis.
1.3 To exploit a multi-modal deep learning strategy to predict glaucoma onset and progression combing clinical, genetic and metabolomics data.

2. Scientific rationale:
Glaucoma is the leading cause of irreversible blindness worldwide.It is a multifactorial disease, whith intraocular pressure (IOP) being the only modifiable risk factor. However, for many glaucoma patients, the disease progresses even with a well-controlled IOP, suggesting that there are other factors contributing to glaucoma. Additionally, the onset and progression of glaucoma is insidious. Patients are aware of the disease until reaching the advanced stages. Thus, it is urgent to recognize the early markers for the disease. Our team is working on a multi-modal deep learning strategy using clinical data, OCT and fundus photography to recognize and grade the severity of glaucomatous visual field damages. Recent GWAS and metabolomics studies have also shown the possible genetic and metabolomic mechanisms of glaucoma. Therefore, combing the clinical, genetic and metabolomics data may help identify novel strategies for predicting.As a result, we will better manage glaucoma beyond controlling eye pressure.

3. Project duration
The project will last 36 months.

4. Public health impact
The UK Biobank dataset comprises a vast number of participants, allowing for robust statistical analyses with increased statistical power to detect associations.Using multi-modal strategies to combing clinical data, genetic and metabolomic information, our research can identify biomarkers and risk factors that allow for earlier detection and intervention, enabling timely treatment to prevent glaucoma progression, and personalized treatment plans based on an individual’s genetic and metabolic profile, optimizing IOP-lowering therapy and reducing side effects. Additionally, integration with other omics data from the UK Biobank could provide a holistic view of glaucoma’s molecular underpinnings for better understanding of the disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-multi-omics-and-imaging-based-modeling-study-of-biological-age-prediction-in-the-eye-and-whole-body

A multi-omics and imaging-based modeling study of biological age prediction in the eye and whole body

Last updated:
ID:
808765
Start date:
19 August 2025
Project status:
Current
Principal investigator:
Professor Hanruo Liu
Lead institution:
Beijng Tongren Hospital, Capital Medical University, China

Early prediction and diagnosis of eye and brain diseases as non-renewable organ-related diseases remain challenging. Although techniques such as retinal imaging have been shown to correlate with pathological processes in the brain, current prediction accuracy and generalizability are limited and further validation is urgently needed. Biological age is an important indicator for assessing the aging process, and the wealth of genetic, proteomic, phenotypic and imaging data in UK Biobank offers the possibility of calculating the aging process in individuals. However, existing studies are still limited in the selection and cross-organizational applicability of biological age metrics.
Therefore, this project intends to integrate multi-omics data (including proteomic, genomic, imaging and clinical information) in UKB, construct an individual aging degree assessment model based on machine learning, and explore its potential application in early prediction of ocular and neurodegenerative diseases. We will combine phenotypic and histological features to identify key biomarkers and systematically analyze their commonalities and specificities in different disease types, providing a theoretical basis for understanding aging mechanisms, disease prediction and individualized intervention.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-multi-omics-approach-for-understanding-the-impact-of-the-triple-burden-of-malnutrition-on-the-risk-of-cancer-and-cardiometabolic-diseases-in-the-uk-biobank

A multi-omics approach for understanding the impact of the triple burden of malnutrition on the risk of cancer and cardiometabolic diseases in the UK Biobank

Last updated:
ID:
842740
Start date:
6 October 2025
Project status:
Current
Principal investigator:
Mr Masrie Getnet Abate
Lead institution:
Ghent University, Belgium

The main aim of this research is to comprehensively investigate the profound impact of the triple burden of malnutrition (TBM) on the risk of cancer and cardiometabolic diseases (CMDs). Furthermore, the effects of selection bias adjustment for predicting cancer and CMDs risk will be investigated. Accordingly, the research questions and aims are set as follows:
Research Question(s) and Aim(s)
Questions:
* How will the selection bias adjustment affect the prediction of the risk of cancer and CMDs?
* How do the different forms of malnutrition (undernutrition, overnutrition, and micronutrient deficiencies) impact cancer and CMDs?
* What are the mediating effects of the multi-omics (proteomic and metabolomic) factors linking the triple burden of malnutrition with cancer and CMDs?

Aims:
* Assess the effects of selection bias adjustment for the prediction of the risk of cancer and CMDs.
* To compare the impact of different forms of malnutrition on the risk of cancer and CMDs.
* To assess the mediating effects of multi-omics factors linking the TBM with the risk of cancer and CMDs.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-multi-omics-data-integration-approach-for-precision-medicine-and-improved-clinical-trial-success

A Multi-omics Data Integration Approach for Precision Medicine and Improved Clinical Trial Success

Last updated:
ID:
57686
Start date:
17 February 2020
Project status:
Closed
Principal investigator:
Professor Sorin Draghici
Lead institution:
AdvaitaBio Corporation, United States of America

This project will develop a novel analysis method and software package able to identify subtypes of disease based on the integration of multiple types of omics data. Many drug candidates fail and many patients receive inappropriate treatment because of our current inability to distinguish between subgroups of patients (respondent vs. non-respondents) and/or subtypes of disease (aggressive vs. non-aggressive). We are developing an approach that will be used both to optimize treatment by separating patients with aggressive disease from those with less aggressive disease, and to increase the success of clinical trials by sub-typing patient groups that are more likely to be respondents from those non-respondents.

Our algorithm will first be used to sub-type patients based on variant data only, then clinical data and omics data will be incorporated to improve the sensitivity and specificity of the sub-typing. UK Biobank is a critical asset for this research because it offers high quality omics and clinical data from a very large population sample. After fine-tuning our algorithm and establishing its effectiveness based on UK Biobank data, we will replicate the research in an independent dataset derived from the US ClinicalTrials.gov database.

We anticipate that this research will significantly increase the effectiveness of clinical trials, and make a contribution to the field consistent with peer-reviewed publication. The proposed work is scheduled for a total of two years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-multi-omics-investigation-of-risk-factors-contributing-to-the-health-disparities-in-south-asians-and-white-europeans

A Multi-Omics Investigation of Risk Factors Contributing to the Health Disparities in South Asians and White Europeans

Last updated:
ID:
490293
Start date:
17 July 2025
Project status:
Current
Principal investigator:
Dr Weihua Zhang
Lead institution:
Imperial College London, Great Britain

South Asians have a 2- to 3-fold higher risk of type 2 diabetes and coronary heart disease than White Europeans but may exhibit lower all-cause and cancer mortality (1,2). The reasons for these disparities remain unclear. Omics data-covering metabolomics, proteomics, and whole-genome sequencing-offer a robust lens to uncover the biological mechanisms behind these differences.
Data Sources
1. South Asia Biobank – ~100,000 South Asian participants in the UK, with up to 20 years of follow-up on lifestyle, environment, sociodemographic, clinical markers, genetics, and metabolomics (3).
2. UK Biobank – A large dataset including White Europeans and ~8,000 South Asians, with over 15 years of follow-up.
Research Questions
1. Which lifestyle, genetic, and omics factors best predict cardiovascular disease, type 2 diabetes, cancer, and all-cause mortality etc.?
2. How do these risk profiles differ between South Asians and White Europeans?
3. Can these findings be generalized or adapted to guide disease prevention in other ethnic groups?
Objectives
1. Integrate data from both cohorts to enhance statistical power and capture a broad range of genetic, environmental, and lifestyle factors.
2. Identify and validate key risk factors (e.g., lifestyle, polygenic risk scores, metabolic, and proteomic profiles) associated with disease in each ethnic group.
3. Compare risk factor contributions between South Asians and White Europeans to uncover the main drivers of observed health disparities.
Expected Impact
By clarifying how various risk factors shape disease trajectories, this research will inform targeted prevention strategies and personalized clinical interventions. Although focused on South Asians and White Europeans, insights gained may extend to other populations and guide broader public health measures.
References
1. Gholap N, et al. Primary Care Diabetes. 2011;5(2):45-56.
2. Bhopal RS, et al. PLoS Med. 2018;15(3):e1002515.
3. https://www.sabiobank.org/


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-multi-omics-perspective-on-eye-disease-prediction

A Multi-Omics Perspective on Eye Disease Prediction

Last updated:
ID:
515707
Start date:
22 May 2025
Project status:
Current
Principal investigator:
Professor Jian Wu
Lead institution:
Henan Academy of Innovations in Medical Science, China

With the global population aging, the prevention and control of chronic diseases have become a major challenge. Due to the non-invasive nature of ophthalmic examinations, the eye serves as a crucial window for assessing overall health and detecting systemic diseases. Traditional research has established associations between ocular diseases or specific ocular parameters and multi-omics data.

Recent advancements in artificial intelligence have revolutionized the extraction and analysis of large-scale molecular data. High-dimensional data from various omics disciplines are increasingly accessible, and the integration of different omics data is gaining attention. Multi-level and multi-factor analyses based on genomics, transcriptomics, proteomics, and metabolomics provide a comprehensive perspective on the molecular mechanisms driving disease occurrence and progression. This study aims to elucidate the multifactorial nature of common eye diseases and, by integrating multi-omics with ocular health, identify novel ocular biomarkers for early disease diagnosis, pinpoint potential therapeutic targets, and formulate public health strategies for the prevention and management of chronic eye diseases.

This study aims to deepen the understanding of the complex interactions between multi-omics and ocular health, elucidating the mechanisms linking the two. These insights are essential for developing individualized medical treatments and improving health outcomes across diverse populations.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-multi-omics-strategy-to-characterize-the-causes-of-cardio-and-cerebrovascular-diseases

A multi-omics strategy to characterize the causes of cardio- and cerebrovascular diseases

Last updated:
ID:
62518
Start date:
29 January 2021
Project status:
Current
Principal investigator:
Professor Guillaume Lettre
Lead institution:
Montreal Heart Institute, Canada

A) Rationale. Healthy blood vessels are critical to prevent the development of diseases such as heart attack, stroke and dementia, which have an important economical and societal burden in the world. Many risk factors (e.g. obesity, high blood pressure) affect both these diseases of the heart and the brain. In this project, we aim to better understand the different genetic and environmental causes of heart and brain diseases characterized by unhealthy blood vessels.

B) Question and Aims. We want to understand why some individuals suffer from heart and brain diseases, whereas others don’t. In particular, our project will help us identify specific genes which contribute to disease risk (or protection). We will combine this genetic information with other environmental variables (e.g. diet, exercise) and clinical parameters (e.g. cholesterol levels) to develop methods that can predict individuals at higher risk of disease. We will also use the genetic information to better understand what molecules are responsible for disease risk; these molecules could become interesting drug targets.

C) Project duration. This project will last three years.

D) Impact. Heart and brain diseases caused by unhealthy blood vessels are responsible for an important morbidity and mortality. We expect that our project will generate tools to predict disease risk and will help guide the development of more efficacious drug targets.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-multi-omics-study-of-gastrointestinal-diseases-based-on-uk-biobank-data-from-mechanisms-to-therapeutics

A Multi-Omics Study of Gastrointestinal Diseases Based on UK Biobank Data: From Mechanisms to Therapeutics

Last updated:
ID:
980679
Start date:
19 August 2025
Project status:
Current
Principal investigator:
Miss Qionglin Huang
Lead institution:
Zhujiang Hospital of the Southern Medical University, China

Research Question & Objectives
Gastrointestinal diseases-colorectal cancer, IBD, NAFLD, peptic ulcer-are rising public-health burdens. Genetic and environmental drivers are known, yet how environmental exposures, psychosocial stressors, and multi-omic profiles interact remains unclear. Using UK Biobank, we will (1) discover novel biomarkers via integrated omics-environment-psychosocial analyses; (2) clarify causal links among exposures, molecular changes, and disease progression; (3) build machine-learning models for early risk prediction and targeted prevention.

Scientific Rationale
Colorectal cancer is a leading cause of cancer death, shaped by genetics, lifestyle, and metabolism. Radiotherapy for pelvic cancers frequently causes chronic radiation-induced intestinal injury. Despite distinct etiology, it shares inflammation and immune dysregulation with other GI diseases, offering a cross-disease model. Circulating proteins, metabolites, and imaging biomarkers illuminate mechanisms. Chronic stress and depressive symptoms amplify gut inflammation and are amplified by immune-metabolic imbalance. Environmental stressors-air pollution, poor diet, sedentary behavior-modulate the gut-brain axis via microbiota, immunity, and neuroendocrine pathways. Most studies examine single omics or isolated exposures, ignoring bidirectional psychosocial interactions. Integrating genomics, proteomics, metabolomics, high-resolution environmental data, mental-health assessments, and accelerometry-derived physical activity will reveal new pathways, refine risk stratification, and guide precision prevention. Advanced causal-inference and machine-learning tools will ensure robust, predictive models ready for translation.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-multi-polygenic-score-of-clock-resilience-and-its-utility-in-predicting-vulnerability-to-night-work-with-regards-to-chronic-disease-risk-cardiovascular-disease-type-2-diabetes-depression-breast

A multi-polygenic score of clock resilience and its utility in predicting vulnerability to night work with regards to chronic disease risk (cardiovascular disease, type 2 diabetes, depression, breast)

Last updated:
ID:
48576
Start date:
30 July 2019
Project status:
Current
Principal investigator:
Professor Eva Susanna Schernhammer
Lead institution:
Medical University of Vienna, Austria

Virtually every cell of our body follows the 24-hr circadian rhythm of a hypothalamic master pacemaker that evolved in the natural light-dark cycle. Decoding this biologic clock culminated in the Nobel Prize in Physiology or Medicine 2017 for the discovery of molecular mechanisms controlling the circadian rhythm. It is now recognized that a strong, unperturbed biologic clock is a hallmark of healthy aging. The introduction of electric light, however, presents unique challenges: today, 20% of the global work force engages in alternate working hours associated with light in unnatural times (eg. night work). Increases in the risk of major chronic disease and mortality have been associated with night work. Further, the ubiquity of light at night implicates potential risk for everyone.
This 5-year project targets the urgency of preventing adverse health consequences of a challenged clock. It does so by aiming to decipher individual risk and related mechanisms 1) using a cutting-edge multi-polygenic score approach; 2) employing transgenerational and deeply phenotyped cohort approaches; and 3) carrying out interventions using genetic risk stratification.
The ultimate aims of this project are to 1) identify the night worker who develops disease, 2) profile mechanisms involved, and 3) optimize effectiveness of interventions to improve sleep and shift work disorder, facilitating immediate implementation of far reaching risk-based prevention strategies/policies, aimed at promoting healthy aging despite clock challenges.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-multidimensional-assessment-of-residual-cardiovascular-risk-in-patients-with-well-controlled-dyslipidemia-an-integrated-study-of-diet-proteomics-metabolomics-and-genetics

A Multidimensional Assessment of Residual Cardiovascular Risk in Patients with Well-Controlled Dyslipidemia: An Integrated Study of Diet, Proteomics, Metabolomics, and Genetics

Last updated:
ID:
267212
Start date:
4 February 2025
Project status:
Current
Principal investigator:
Dr Fenghua Ding
Lead institution:
Ruijin Hospital, China

Aims:
Our project aims to uncover the lingering risks of cardiovascular diseases in people who have managed to get their cholesterol and other blood fats to healthy levels. We want to find out why some people still face health issues related to heart diseases even when their lipid levels are under control.
Scientific Rationale:
Cardiovascular diseases are a major cause of death and disability worldwide. High cholesterol and other lipid abnormalities are key risk factors, but even when these are well-managed, there’s still a risk. We suspect this leftover risk could be due to factors like diet, genes, and how the body processes certain substances. New technologies are giving us a chance to look at this in more detail.
Project Duration:
We plan to spend several years on this research to gather comprehensive data from participants, perform detailed analyses of dietary habits, plasma proteomics, metabolites, and polygenic risk scores, and then construct and validate a risk assessment model.
Public Health Impact:
Through this study, we aim to gain a more comprehensive understanding of the residual cardiovascular risk in patients with well-controlled dyslipidemia. By providing more precise risk assessment and management strategies for clinical practice, we hope to ultimately improve patient outcomes and quality of life. This research will contribute to the development of personalized medicine approaches, tailoring interventions to individual patient profiles, and enhancing the effectiveness of preventive measures against cardiovascular diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-multifactorial-investigation-into-the-determinants-and-outcomes-of-sleep-and-circadian-health

A multifactorial investigation into the determinants and outcomes of sleep and circadian health

Last updated:
ID:
93538
Start date:
7 November 2022
Project status:
Current
Principal investigator:
Dr Eva C Winnebeck
Lead institution:
University of Surrey, Great Britain

Sleep is not a commodity but an essential biological process. Good quality and sufficient sleep helps to maintain health, well-being and performance, while poor sleep has been found to put people at risk, both in the short-term (acute effects) and long-term (chronic effects). But what makes people sleep better or worse, later or earlier, more or less, regularly or irregularly? And which of these sleep aspects influence which aspects of health?

In this project, we seek to use the extensive data of the UK Biobank to shed more light on the factors that can influence sleep as well as the possible consequences for health. The factors that we will study are individual factors such as participants’ genetics and chronotype, environmental factors such as season or weather as well as sleep factors such as how and when an individual slept the night before. To assess potential health consequences, we will be investigating the relationship between various sleep features and common diseases that affect large parts of the population e.g. diabetes, atherosclerosis or depression.

We aim to expand on previous findings by deriving new measures of sleep habits and circadian variability, primarily through accelerometer data, investigating the contribution of genetics on new and existing sleep measures through analysis of genotyping or sequence data and explore the long-term effects of sleep and circadian dysfunction on disease risk and progression by incorporating information on genetic predisposition with ongoing healthcare records. We intend to build a more complete picture of how a combination of genetics and environment (and their interaction) contribute to the range of sleep habits (and dysfunction) seen amongst the population.
The project is planned to be long-lived, incorporating new methodology, data sources and expertise as they arise. The initial period will be three years. The insights from our project should aid both basic research and public health. A better understanding of the importance of sleep for healthy living and better knowledge about which factors lead to poor sleep enables better public health campaigns and prioritisation for preventive medicine to improve population health. Insights into the genetics of sleep patterns can help to identify new sleep medicines. Finally, the development of new analysis tools, incorporation of external environmental data and extraction of novel sleep measures will benefit the wider research community and accelerate sleep research.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-multimodal-data-driven-study-on-the-clinical-and-mechanistic-associations-between-lifes-essential-9-with-sarcopenia-frailty-and-age-related-cardiovascular-diseases

A Multimodal Data-Driven Study on the Clinical and Mechanistic Associations Between Life’s Essential 9 with Sarcopenia, Frailty, and Age-Related Cardiovascular Diseases

Last updated:
ID:
862905
Start date:
18 September 2025
Project status:
Current
Principal investigator:
Dr Xiaoyao Li
Lead institution:
Fuwai Hospital Chinese Academy of Medical Sciences, China

Research Objective
This study aims to evaluate the associations between Life’s Essential 9 (LE9) and sarcopenia, frailty, and age-related cardiovascular diseases (CVDs) in the UK Biobank (UKB) cohort.
Scientific Rationale
With global aging, sarcopenia, frailty, and CVDs have become interrelated conditions that significantly affect older adults. In 2024, Circulation proposed LE9-a comprehensive framework including four behavioral metrics (diet, physical activity, nicotine exposure, sleep) and five health factors (BMI, blood pressure, blood glucose, blood lipids, and psychological health)-as an updated measure of cardiovascular health (CVH).
Sarcopenia is not only a marker of physical decline but also associated with CVDs. few studies have examined how LE9 components, especially modifiable factors like diet, physical activity, sleep, and mental health, interact with sarcopenia, frailty, and age-related CVDs (e.g., coronary artery disease, arrhythmia, heart failure). It remains unclear whether optimal CVH can offset genetic or environmental risks.
We aim to utilize the entire UK Biobank cohort, including data on socioeconomic status, lifestyle and environmental factors, genomics (such as aging-related clonal hematopoiesis [CHIP], sarcopenia-related whole exome sequencing, whole genome sequencing, and genotyping), biochemical markers, and health outcomes (death/cancer registries, cardiometabolic diseases, and first occurrences). We will also incorporate online follow-up data (24-hour dietary recalls, cognitive function assessments) and imaging data. We will apply genetic risk scores (GRS) and deep learning methods to assess associations between LE9 and aging-related CVD and explore the underlying biological mechanisms within genetic contexts.
Public Health Significance
This study will show LE9’s value in predicting sarcopenia/frailty and CVD, and reveal interactions between CVH and genetic risk. Findings will promote healthy aging.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-multiscale-genotype-phenotype-map

A multiscale genotype – phenotype map

Last updated:
ID:
54343
Start date:
30 October 2019
Project status:
Closed
Principal investigator:
Dr Eduard Porta Pardo
Lead institution:
Barcelona Supercomputing Center, Spain

Genetic variants can be classified into two different groups according to whether they affect the parts of the genome that tell cells how to make proteins or not. The former are called coding mutations, and they can have large effects in an individual. The latter are called non-coding mutations and their effects are usually more modest. So far, we have identified hundreds of non-coding variants associated to higher or lower cancer risk, but we have had limited success in finding coding variants.

We believe that our limited success with coding variants is due to the way we analyze them. So far, we either look at the effect of each coding variant on its own, or we group them according to the protein they affect. However, proteins have different parts with different functions, and the effect of each coding variant likely depends on which precise part of the protein it affects. We have recently developed a new computational tool that uses the information of the different parts of the protein to group the genetic coding variants. We will use this new tool to identify new protein regions that are associated with higher or lower cancer risk.

Finally, we will develop an artificial intelligence model that integrates the effects of both, the coding and non-coding variants of each individual, to predict his or her risk of suffering cancer in the future. If successful, this model could help physicians decide whether a person has a high or low risk of cancer. This is extremely important, as people with high risk should be screened more often, whereas people with low risk might be able to avoid some screens and the discomfort that they might have associated.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-neurobiological-and-genetic-study-of-loneliness

A neurobiological and genetic study of loneliness

Last updated:
ID:
68324
Start date:
28 January 2021
Project status:
Closed
Principal investigator:
Dr Richard Ramsey
Lead institution:
Macquarie University, Australia

Loneliness is an increasingly common experience found in the general population, especially in younger adults and the elderly. The experience of loneliness has detrimental outcomes for mental health as well as physical health, with much research showing links between loneliness and both disease and premature death. The effects of loneliness on health and wellbeing are so severe that its associated morbidity and mortality rates are comparable to that of smoking and obesity.

The proposed research project aims to investigate the associations and possible causes of loneliness, taking into account psychological and social factors, as well as differences in the brain, biology and genetics. We hope that by understanding how loneliness appears in the population, we may be able to gain an insight into how we can reduce experiences of loneliness and possibly even treat loneliness in the future. The project will take place over a period of 3 years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-new-intervention-for-the-implementation-of-pharmacogenetics-in-psychiatry-psy-pgx

A new intervention for the implementation of pharmacogenetics in Psychiatry (PSY-PGx)

Last updated:
ID:
92773
Start date:
28 February 2024
Project status:
Current
Principal investigator:
Dr Roos van Westrhenen
Lead institution:
Parnassia Psychiatric Institute, Netherlands

Recent evidence suggests that genetic variations in the proteins that facilitate drug metabolism can identify which patients benefit from which treatment. This translates to a reduction in side effects and better efficacy of medication, thereby reducing the suffering for the individual patient and quicker recovery. This proposed work will determine which individual patient characteristics, including pharmacogenetics, influence medication response, allowing for the construction of novel models and algorithms for personalized medication prescription. This will include an initial investigation of pharmacogenes, genes that are known to have some impact on medication response. The large datasets that comprise the UK Biobank are a key component to the development of this algorithm and will be used in conjunction with big data from the Finnish Biobank as part of the larger PSY-PGx project (https://www.psy-pgx.org/PSY-PGx). Altogether, we anticipate the project lasting 60 months. Ultimately, this project tackles the expected growth in mental illness in the EU, benefit millions of patients and make for a healthier population leading to cost reductions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-novel-approach-integrating-genomics-into-the-health-economic-modelling-of-therapeutics

A novel approach integrating genomics into the health economic modelling of therapeutics

Last updated:
ID:
88775
Start date:
13 July 2022
Project status:
Current
Principal investigator:
Professor Zanfina Ademi
Lead institution:
Monash University, Australia

For people at risk of chronic disease, it is often better to start treatment early to prevent or delay the disease altogether, rather than the all-too-common approach of waiting until overt disease develops before intervening. However, even if at high risk of the disease of their lifetime, it is often difficult for younger and middle-aged people without overt disease to access newer therapeutics. This is because treatments only become widely available when shown to be cost-effective, and current health economic models (which underpin these cost-effectiveness analyses) often do not account for the high lifetime risk of chronic disease in some individuals.
Genetic risk scores can now predict the long-term risk of chronic disease from birth, and thus it is possible to identify individuals most likely to benefit from early intervention to prevent disease. In this project, we will use genetic risk scores to identify populations at high lifetime risk of chronic disease, and evaluate the costs and benefits of targeting health interventions to them early in life, before their chronic disease develops.
This will involve first assessing which individuals, by virtue of their genome, are likely to benefit most from certain interventions (for example, some individuals have genetically high cholesterol levels that predispose them to heart disease and are thus likely to benefit most from early cholesterol-lowering treatments). We can then estimate the lifetime risk of chronic disease in these individuals. These estimates then form the basis of our health economic model. We can then use this model to project the costs and benefit of intervening (compared to not intervening).
Unfortunately, due to limited healthcare resources, just proving benefit of an intervention does not guarantee that payers (the government and/or health insurers) will make this intervention available, particularly when the intervention is expensive. Payers usually require evidence of cost-effectiveness to provide access to interventions. Therefore, the key outcome of this project will be to develop a new framework for how new interventions are made available, or how existing interventions targeted to new populations, with the ultimate aim of increasing access to healthcare in a cost-effective manner.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-novel-integrated-predictive-model-for-pancreatic-cancer-in-the-uk-biobank-cohort

A novel integrated predictive model for pancreatic cancer in the UK Biobank cohort.

Last updated:
ID:
94611
Start date:
30 November 2022
Project status:
Current
Principal investigator:
Dr Temin Ke
Lead institution:
University of Manchester, Great Britain

Globally, pancreatic cancer (PaCa) is the 12th most common cancer and the 7th leading cause of cancer death. In the UK, PaCa is the 10th most common cancer, with approximately 10,449 people diagnosed annually (year 2016-2018). Unlike the apparent improving survival rates for common cancers such as breast and colorectal cancer, not much progress in terms of survival rate has been made for PaCa. The 1-year survival rate of PaCa is approximately 28%, and the 5-year survival rate is about 6%. However, Cancer Research UK(CRUK) has reported that 37% of pancreatic cancer patients in the UK can be prevented. Due to the emerging increased incidence and poor prognosis of PaCa, it is crucial to confirm the PaCa factors and target people at risk in the UK population.

Previously published literature has reported many potential PaCa risk factors; however, there were some inconsistent findings. Some probable risk factors include ageing, male gender, African American ethnicity, cigarette smoking, heavy alcohol consumption, increased body mass index (BMI), abdominal obesity, chronic pancreatitis, Diabetes mellitus (DM), hepatitis B and cholecystectomy, family history, germline mutation, ABO blood type and genetic predisposition, etc. Some inconclusive potential risk factors include low physical activity, increased consumption of red/processed meat and dairy products, Vitamin D insufficiency, Helicobacter pylori (H. pylori) infection, long-term use of Proton-pump inhibitors(PPI), and Systemic Lupus Erythematosus(SLE) and anti-diabetic drugs, etc. On the other hand, there is a lack of evidence from a large population-based cohort study. Moreover, there was no comprehensive predictive model to combine the lifestyle-related factors, medical-related factors and genetic disposition in the previous survey.

This study would like to investigate the comprehensive PaCa risk factors through the UK Biobank cohort. The UK Biobank is a large population-based prospective cohort study that collected data from around half a million UK people. The recruiting was carried out from 22 centres to ensure that population coverage was dispersed across the UK. This study aims to explore potential risk factors, including lifestyle-related risk factors, medical-related risk factors and genetic predisposition in the UK population. Furthermore, a comprehensive predictive model will be established based on these risk factors, which may provide a reference for clinicians and researchers for the following pancreatic cancer-related risk stratification, prevention, and early detection studies. The findings could also be used as a reference to target people at risk of pancreatic cancer in the UK community to promote the pancreatic cancer prevention program.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-novel-reward-system-polygenic-risk-score-for-the-prediction-of-obesity-and-substance-use-disorders

A Novel Reward System Polygenic Risk Score for the Prediction of Obesity and Substance Use Disorders

Last updated:
ID:
48083
Start date:
27 March 2019
Project status:
Current
Principal investigator:
Dr Shannon K McWeeney
Lead institution:
Oregon Health & Science University, United States of America

We currently do not know if there is a shared genetic risk for adult-onset obesity and substance use disorders. The aims of this research project are: (1) to uncover any genetic risk factors associated with reward system function, including palatable food intake and substance use, and (2) to use these genetic risk factors to improve the prediction of obesity and substance use disorders in the clinic. By investigating a common genetic signal for the reward system, this research project may improve prediction of other adult-onset complex diseases. The duration of this project will last approximately 18 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-novel-statistical-approach-to-tackle-the-current-methodological-problems-associated-with-genotype-by-environment-interactions

A novel statistical approach to tackle the current methodological problems associated with genotype-by-environment interactions.

Last updated:
ID:
720967
Start date:
11 July 2025
Project status:
Current
Principal investigator:
Dr Sang Hong Lee
Lead institution:
University of South Australia, Australia

Complex diseases and traits arise from the interplay of genetic and environmental factors. Understanding these interactions is crucial, yet current statistical methods for genotype-by-environment (GxE) interaction analysis often fail to accurately determine causal directions. This project proposes the development of a novel framework, the Genetic Causality Inference Model (GCIM), which infers causal relationships without assuming a predefined direction. Many existing methods rely on prior knowledge of the causal direction in GxE interactions, and if these assumptions are incorrect, they can lead to misleading conclusions.
The primary objective of this project is to develop GCIM to accurately determine the causal directions of GxE interactions. The model will be validated through simulations with varying scenarios, and its performance will be compared to that of existing GxE methods. The model will also be applied to UK Biobank data, which offers comprehensive phenotypic, genetic, and environmental data essential for robust GxE analyses. This dataset will allow us to analyze the causal directions of complex traits and diseases with rigorous quality control measures and confounding adjustments to ensure the reliability of our findings.
This project seeks to address a common limitation of current GxE methods: the potential for misinterpretation when the true causal direction is unknown. GCIM overcomes this by inferring causal directions without predefined assumptions. Through simulations and application to UK Biobank data, the proposed method will be thoroughly validated and verified. The validated method will enhance GxE analysis in complex diseases. The developed framework will be integrated into user-friendly software and made publicly available. The findings will be disseminated through high-impact publications and scientific platforms, promoting collaboration and contributing to personalized medicine by incorporating individual environmental exposures.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-phenome-wide-association-analysis-and-mendelian-randomisation-analysis-on-metabolic-features-detected-by-untargetted-metabolomics

A phenome-wide association analysis and Mendelian Randomisation analysis on metabolic features detected by untargetted metabolomics

Last updated:
ID:
52569
Start date:
21 July 2020
Project status:
Current
Principal investigator:
Dr Abbas Dehghan
Lead institution:
Imperial College London, Great Britain

Advanced metabolomics methods have now made it possible to measure thousands of small molecules in body specimen including blood. These molecules are important since they could inform us regarding human health and could in some instances be used as target for prevention or treatment of diseases. Although many studies have investigated the relation of the blood levels of these molecules with diseases, its not clear whether the relation is causal. A third factor might be driving the relation or the blood levels of the metabolite might have been afected by the early stages of the disease. Assessment of the causal effect is normally done in a trial setting where participants receive – in random – medications of other interventions that affect the blood levels of the metabolite. However, specific interventions for these molecules are not known and in many instances such studies are not ethical. An alternative approach is to use genetic information (so called Mendelian Randomisation approach). In this approach, genetic variants that are related to blood levels of the metabolite are studied for their relation with clinical traits and disorders. Given that genetic variants are inheritred in random, any differences in the comparison group should be due to the causal effect of the metabilte.

In another projedct, we have identified genetic variants for a wide range of metabolies and are interested to test their associations with traits and diseases. In this project, we wil use data from UK bio bank to study the association of the genetic variants with a wide range of clinical traits and diseases. This will unravel the clinical importance of the studied metabolites in an agnostic approach and the results could be used either to build or improve risk prediction models or provide novel drug target candidates.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-phenome-wide-association-and-mendelian-randomisation-study-of-polygenic-risk-for-alzheimers-traits-in-uk-biobank

A phenome-wide association and Mendelian Randomisation study of polygenic risk for Alzheimer’s traits in UK Biobank

Last updated:
ID:
84254
Start date:
6 December 2022
Project status:
Current
Principal investigator:
Professor Jintai Yu
Lead institution:
Fudan University, China

In the entire world, Alzheimer’s disease is the number one cause of morbidity and mortality. The need for primary prevention has been reinforced by a clear decline in dementia prevalence and incidence that has been linked to earlier population-level investments like better vascular health and education. However, only a small number of modifiable risk factors were found, and if they were properly managed, they only reduced the risk of disease by 40%. There are still many unidentified risk factors. In this study, we seek to identify novel environmental factors that can be changed that influence the onset of Alzheimer’s disease and to identify potential targets for interventions. To be more precise, we will 1) evaluate the impact of various environmental risk factors, such as lifestyles, comorbidity, medications, local environmental exposures, etc.; 2) investigate the potential interaction between these different factors; 3) identify people with genetic susceptibility and the way effect of risk factors can be modified by genetic variation; and 4) assess whether lifestyle factors play a role as potential effect modulator. A better understanding of the interplay between different modifiable environmental factors and the development of Alzheimer’s disease is expected to provide new evidence for intervention target and facilitate more efficient prevention strategies and. This project may lead to a more holistic approach to personalized prevention and treatment, accounting for both individual-level factors and community-level environmental factors.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-phenome-wide-association-and-mendelian-randomization-study-for-parkinsons-disease

A Phenome-wide Association and Mendelian Randomization Study for Parkinson’s disease

Last updated:
ID:
104811
Start date:
23 June 2023
Project status:
Current
Principal investigator:
Dr Changhe Shi
Lead institution:
First Affiliated Hospital of Zhengzhou University, China

The aim of this study was to systematically screen and validate a wide range of potential risk factors for Parkinson’s disease (PD). Current studies have found that the incidence of PD is closely related to many factors, among which genetic factors play an important role. Large-scale Genome wide association study (GWAS) analysis further confirmed the role of gene variation in PD incidence. However, current studies mainly discuss the relationship between PD and risk factors by setting risk factors, and many risk factors are still ignored or unknown. The phenome-wide association study (PheWAS) are hypothesis-free analyses that examine genetic associations of a wide range of factors with disease. In view of the fact that genetic association can be used as a tool to identify risk factors for PD, this study intends to apply the polygenic risk score (PRS) of PD to represent the risk of PD, which can systematically screen a wide range of established and unknown non-genetic factors associated with PD. On this basis, two-sample Mendelian randomization and brain structures association analysis were used to further evaluate the potential causal relationship between the identified factors and PD and explore the potential biological mechanism of PD-PRS and PD-related factors. During this study, which is expected to be completed within 12 months, a comprehensive and rigorous assessment of the association between a wide range of risk factors and PD will be conducted. The systematic integration of genetic, clinical and neuroimaging information will help to identify risk factors and prevention goals for PD.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-phenome-wide-association-study-for-cardiac-diseases-using-mendelian-randomization-mr-and-artificial-intelligence-ai

A phenome-wide association study for cardiac diseases using Mendelian randomization (MR) and Artificial intelligence (AI).

Last updated:
ID:
81679
Start date:
20 April 2022
Project status:
Current
Principal investigator:
Professor Hui-Nam Pak
Lead institution:
Yonsei University, Korea (South)

Cardiovascular diseases have been known to associate with genetic predisposition, however, the pleiotropic effects of a single genetic variant or group of genetic variants on various disease phenotypes have not been well understood. Phenome-wide association studies (PheWAS) are defined as a statistical approach to test for the association of a genetic variant or accumulative effects of genetic variants and a wide range of phenotypes. PheWAS has been widely used as the large genetic data set has recently become available. PheWAS is particularly useful in situations where we currently have an incomplete understanding of disease mechanisms. Moreover, PheWAS approaches were based on genotypes that are fixed from birth, so it is less affected by environmental confounding factors and reverse causality. This research project aims to investigate the phenotypes that were associated with a polygenic risk score for cardiovascular diseases, and to study whether there is a causal association of cardiovascular disease-polygenic risk score (PRS) and the phenotypes using mendelian randomization (MR). MR is a widely used epidemiological method that utilizes genetic variants to investigate casual association of a risk factor and an outcome. In MR, genetic variants that satisfy all the assumptions are used as an instrumental variable (IV). Assumption of MR includes that genetic variants are associated with risk factors and IV are not directly associated with confounders of the risk factors, nor does it affect the outcome directly. The goal of the research project is to discover the causal risk factors for cardiovascular diseases and to construct a predictive model for new diagnostic tests or treatments. The project duration is about 36 months. Clinical implications include understanding the genetic causes of cardiovascular diseases and applying such information for clinical practice. Public health impacts are concerned with discovering and predicting causative risk factors for cardiovascular diseases that can be a cornerstone of timely clinical management.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-phenome-wide-association-study-of-copy-number-variation

A phenome-wide association study of copy number variation

Last updated:
ID:
17731
Start date:
24 January 2017
Project status:
Current
Principal investigator:
Professor Nathan Pankratz
Lead institution:
University of Minnesota Twin Cities, United States of America

To identify changes in the DNA, particularly large deletions and duplications of the genome, that are related to complex diseases and intermediate biomarkers The identification of genetic risk factors that are related to disease will help reveal the underlying biology and thereby expose targets for environmental modification and/or novel drug development. We will use the raw Affymetrix Axiom data (.CEL files) and apply a principal components analysis on these intensities to correct for DNA quality and batch effects. We will then perform a joint calling across all samples to determine potential deletions and duplications using our soon to be open-source Genvisis software package, which will also assess the quality of these calls and filter them down to a set of high quality calls (~1 year to complete). We will then associate the high quality calls with the phenotypes available, and replicate any findings using external data sources (~1 additional year). Full cohort; all samples with high quality intensity data will be analyzed


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-phenotype-wide-and-omics-analyses-of-social-and-environmental-exposomes-sleep-architectures-aging-related-multimorbidity-with-pre-clinical-brain-pathological-changes-and-incidence-of-dementia

A phenotype-wide and omics analyses of social and environmental exposomes, sleep-architectures, aging related multimorbidity with pre-clinical brain pathological changes and incidence of dementia

Last updated:
ID:
203528
Start date:
30 October 2024
Project status:
Current
Principal investigator:
Jiayuan Li
Lead institution:
West China Fourth Hospital, Sichuan University, China

With the global rise in the elderly population, there has been a notable increase in the occurrence of diseases and mortality rates, with dementia being a major contributor. Dementia not only leads to functional impairments but also reduces life expectancy, making it a critical concern for public health. However, most existing studies on dementia primarily rely on epidemiological analyses, lacking comprehensive pre-clinical brain image features and omics-based analyses that are crucial for understanding the development of early-stage dementia. Additionally, although previous research has explored risk factors associated with dementia, such as diet, physical activity, sleep quality, metabolites, air pollution, and living in greener environments, no systematic studies have been conducted to assess the combined effect of these factors. Meanwhile, the precise mechanism by which these factors influence the risk of dementia, particularly the pre-clinical brain pathological changes, remains largely unclear, making early-stage dementia development elusive. Moreover, while incident multimorbidity, including conditions like obesity, stroke, depression, and sleep disturbances, has been reported to be associated with dementia, validating these associations requires large-scale prospective studies.
Understanding the onset of dementia is of clinical significance as it can aid in prevention and delay the progression of the disease. Therefore, our project aims to (1) examine the independent and joint associations of social, dietary, behavioral, and environmental exposomes with dementia-related brain changes and incidence using prospective cohort studies; (2) uncover the underlying mechanisms of exposomes on dementia risks through omics and genetic analysis; (3) estimate the temporal patterns of multimorbidity leading to dementia transmission, highlighting the relationships and potential mechanisms among exposomes, multimorbidity, and dementia risks. To achieve these objectives, we will conduct a prospective observational analysis, genome-wide association analysis, phenome-wide association analysis, and mendelian randomization analysis. Additionally, a series of secondary analyses will be performed using data from the UK Biobank. Our plan is to begin the analyses as soon as the data become available and aim to complete the project within 36 months, including manuscript preparation for review by authors.
We hope that our study will provide a comprehensive understanding of the associations between exposomes, sleep architectures, aging-related multimorbidity, and pre-clinical brain pathological changes. Additionally, we anticipate identifying novel biological pathways and potential preventive targets for improving dementia prevention. Our project’s objectives align with the goals of the UK Biobank, which is dedicated to enhancing the prevention, diagnosis, and treatment of various serious and life-threatening illnesses, including dementia.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-polygenetic-risk-score-based-analysis-of-gene-drug-interactions-increasing-type-two-diabetes-risk

A Polygenetic Risk Score Based Analysis of Gene-Drug Interactions Increasing Type Two Diabetes Risk.

Last updated:
ID:
94215
Start date:
11 January 2023
Project status:
Closed
Principal investigator:
Dr Luke Wander
Lead institution:
University of Washington, United States of America

It has been shown through many population studies that Statin usage causes an increased risk of developing type two diabetes. Since Statins have been shown to be safe and effective at lowering the risk of cardiovascular disease, they will remain a widely used first line therapy for many people. Therefore, it is clinically relevant to understand the underlying mechanisms of this risk so both preventive and therapeutic actions can be taken.
To explore this risk, we propose to use polygenic risk scores – which describe someone’s genetic predisposition for acquiring a certain disease. Overall, we want to see if those that have a higher genetic risk for certain diseases like diabetes or high cholesterol, have a higher than expected risk of developing new onset type two diabetes when taking Statins.

We have three main aims in this project, the first is to create a model to predict the risk of new onset type 2 diabetes using polygenic risk scores and other factors such as age, BMI and Statin usage. The second is to use this model to test for greater than expected diabetes risk in those taking Statins who have a high polygenetic risk score. And finally, if an interaction exists, test for interactions between single genetic variants that make up the polygenic risk score and Statin usage.

The results of this study may be able to identify what groups are at greater risk for increased diabetes susceptibility related to Statin usage and by what biological pathways this increased risk operates through. Having knowledge of what groups are at a higher risk may enable preventative measures to be taken to reduce this risk. Additionally, knowing what biological pathways this increased risk may be working through could provide insights into therapeutic actions that may be taken to mitigate risk.

This project will take approximately 2 years to complete.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-population-based-study-of-precision-health-with-multi-omics-strategies-and-deep-phenotypes

A population-based study of precision health with multi-omics strategies and deep phenotypes

Last updated:
ID:
202239
Start date:
4 December 2024
Project status:
Current
Principal investigator:
Professor Jintai Yu
Lead institution:
Fudan University, China

Our research project aims to uncover new insights into human diseases and improve patient outcomes by identifying specific markers and potential treatment targets using cutting-edge technologies.
Human diseases, such as heart disease, diabetes, cancer, and neurodegenerative disorders, affect the lives of millions of people worldwide. Despite extensive research, many aspects of these diseases remain unclear. By analyzing a large and diverse dataset from the UK Biobank, which includes genetic, proteomic, metabolomic, and multi-modal imaging information as well as multiple lines of disease information, we will gain a deeper understanding of the complex biological mechanisms underlying these diseases. By studying multiple layers of biological information simultaneously, we can identify common molecular patterns across different chronic diseases and uncover specific biomarkers that can predict disease susceptibility, progression, and response to treatment. The estimated duration of the project is 36 months. The knowledge generated by this project will help in developing personalized and targeted interventions, ultimately improving patient care and reducing the burden of chronic diseases on individuals and society as a whole.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-population-survey-of-subclonal-copy-number-variation

A population survey of subclonal copy number variation

Last updated:
ID:
68245
Start date:
9 February 2021
Project status:
Closed
Principal investigator:
Dr Mark Pinese
Lead institution:
Children's Cancer Institute Australia, Australia

As people age, their blood accumulates genetic changes. One type of change, called “CHIP”, strongly predicts whether a person will soon develop disease such as cancer or heart disease. If we could develop a method to detect CHIP in healthy people, it could revolutionise how these common diseases are managed. Unfortunately, current methods to identify CHIP are slow and expensive, and not ready for application to public health.

We believe we have developed a method to address this, and enable the detection of CHIP using standard techniques that are already used in clinical laboratories. We will use the UK Biobank data to refine and test our methods, and establish whether our measure of CHIP predicts future health in the UK Biobank. This project will take around two years, and if successful has the potential to establish a new approach to identify people at high risk of cancer and heart disease in the population, for targeted surveillance and disease management.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-precision-health-study-based-on-uk-biobank-data

A precision health study based on UK Biobank data

Last updated:
ID:
57561
Start date:
13 February 2020
Project status:
Current
Principal investigator:
Dr Hsin-Chou Yang
Lead institution:
Academia Sinica, Taiwan, Province of China

his study aims to uncover human genome and examine their roles in the promotion of health and welfare. In this study, we will identify genes associated with available clinical phenotypes and quantitative traits in UK Biobank by using genome-wide PheWAS and evaluate how they influence the studied diseases or quantitative traits. This is the first and fundamental step to understand disease etiology and genetic mechanism. Complex diseases are co-influenced by genes, environmental exposures, and their interaction. This study will also examine how disease genes work with environmental exposures to increase a risk of the studied diseases or quantitative traits. The empirical evidence from this study will provide a better understanding about our genome. The information has a potential clinical impact for developing disease prevention, diagnosis and treatment in precision health that different treatments are applied to different patients according to their unique genomic make up. We plan to execute this project for a 3-year period that includes data clean, data analysis, data interpretation, and results presentation and publications.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-precision-medicine-approach-for-treatment-and-prevention-of-alzheimers-disease-using-statins

A PRECISION MEDICINE APPROACH FOR TREATMENT AND PREVENTION OF ALZHEIMER?S DISEASE USING STATINS

Last updated:
ID:
19923
Start date:
1 November 2017
Project status:
Closed
Principal investigator:
Dr Roberta Brinton
Lead institution:
University of Southern California, United States of America

Alzheimer?s disease (AD) has reached global epidemic proportions. Therapeutics to prevent, delay and treat AD are urgently needed.

Significant emerging evidence links cholesterol, Aß and AD, and several studies have shown a reduced risk for AD and dementia in populations treated with statins. The ApoE4 allele of the apolipoprotein E gene, is associated with higher cholesterol levels and increased risk for AD. Preliminary results of our own meta-analysis of clinical trial data indicate that the use of statins may delay or slow down the progression of the disease and cognitive decline, to a greater extent in ApoE4 carriers. Despite substantial research and development investment in Alzheimer?s disease, effective therapeutics remain elusive. Our precision medicine approach will generate scientific evidence needed to drive the development of therapies that treat the right person, with the right treatment at the right time. We propose the use of a precision medicine approach, which takes into account people’s individual variations in genes, environment and lifestyle, for analysis of data from the UK BioBank, in order to further examine the effect that the use of statins may have on the onset and cognitive decline of Alzheimer’s disease. Full cohort


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-predictive-model-for-hematologic-aging-and-the-role-of-lifestyle-in-accelerating-senescence-and-disease-onset

A Predictive Model for Hematologic Aging and the Role of Lifestyle in Accelerating Senescence and Disease Onset

Last updated:
ID:
805798
Start date:
27 June 2025
Project status:
Current
Principal investigator:
Professor changzheng Li
Lead institution:
Guangzhou Medical University, China

Hematopoietic aging represents one of the most critical biological processes impacting global health. Epidemiological studies reveal that approximately 23% of individuals aged 65 or older worldwide exhibit detectable biomarkers of hematopoietic stem cell (HSC) senescence. This population demonstrates a 4.1-fold elevated risk of developing hematologic malignancies-including myelodysplastic syndromes and lymphomas-compared to healthy aging cohorts. According to World Health Organization statistics, complications associated with hematopoietic aging (e.g., thromboembolic disorders, septic shock) account for 18% of mortality causes in elderly populations globally, demonstrating clinical significance comparable to cardiovascular diseases.

Molecular alterations such as leukocyte telomere attrition and epigenetic dysregulation in hematopoietic stem cells (HSCs) may precede overt clinical symptoms by two decades. However, current diagnostic protocols lack the capacity to detect these early warning signals, leaving a critical gap in preventive healthcare strategies. Current diagnostic frameworks relying on conventional blood tests remain limited to detecting late-stage pathological alterations, with insufficient resolution to identify early aging biomarkers.

Our aim is to leverage the UK Biobank database to integrate whole-genome sequencing, complete blood count parameters, plasma proteomic profiles, metabolomic profiles, leukocyte telomere length measurements, and detailed clinical data. By applying advanced machine learning techniques, we aim to establish a multimodal hematopoietic aging prediction system. Based on this model, we will also analyze how lifestyle factors influence hematopoietic aging trajectories and the progression of hematologic disorders. This model could help in accurating diagnosis of individual hematopoietic aging predisposition and predict the onset and progression of blood-related diseases at the personalized level.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-prospective-multicentre-clinical-study-on-the-application-of-biomarkers-in-disease-through-multi-omics-exploration

A Prospective Multicentre Clinical Study on the Application of Biomarkers in Disease through Multi-Omics Exploration

Last updated:
ID:
535832
Start date:
4 February 2025
Project status:
Current
Principal investigator:
Professor Jian Guan
Lead institution:
Nanfang Hospital, Southern Medical University, China

Cancer and chronic diseases, such as diabetes and cardiovascular conditions, represent major global health burdens, with complex biological interconnections and often shared molecular mechanisms, including genetic mutations, metabolic imbalances, and abnormal protein expression. Biomarkers play a critical role in early diagnosis, prognosis, and therapeutic monitoring, as they reflect physiological states and disease characteristics throughout the onset and progression of these conditions. However, many studies have been limited to observational designs, lacking the prospective research needed to fully realise the clinical potential of biomarkers in diagnosis, prognosis, prediction, and monitoring. Long-term follow-up and large-scale studies are crucial to evaluate the effectiveness of biomarkers in clinical practice, which would substantially enhance their applicability. This prospective, multicentre clinical study aims to establish a multi-omics data analysis platform to systematically identify and validate clinically valuable biomarkers. This approach seeks to improve the accuracy of early diagnosis, predict disease progression, support personalised treatments, and develop predictive and diagnostic models, thus providing a robust scientific basis for precision medicine.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-prospective-study-of-daily-lifestyle-living-customary-and-the-risk-of-breast-cancer-in-the-uk-biobank

A prospective study of daily lifestyle/living customary and the risk of breast cancer in the UK Biobank

Last updated:
ID:
62980
Start date:
16 February 2021
Project status:
Current
Principal investigator:
Zhijun Dai
Lead institution:
Zhejiang University, China

The aim of our project (estimated duration: 2 years) is to investigate the effect of lifestyle on breast cancer risk and prognosis, and to evaluate the efficacy of separated or combined lifestyle interventions on the incidence and prognosis of female breast cancer and further explore the possible mechanisms more and more attention has been paid to the impact of daily lifestyle on the incidence and prognosis of breast cancer. Breast cancer is a complex disease resulting from a combination of genetic and environmental factors. Daily lifestyles were reported to be related to cancer risk and prognosis, including physical activity, electronic device use, sleep, smoking, diet habit, alcohol intake and sexual factors et al. Although some studies have explored and speculated on this issue, these researches are not in-depth enough and the results are controversial. Therefore, solid and thorough population researches of this issue are needed. We plan to discuss the lifestyle risk factors effect on breast cancer risk and prognosis separately according to the clinical characteristics, such as menstrual history, age, reproductive history, etc. Lifestyle risk factors can be stratified by severity, such as smoking and alcohol intake, time spent on electronics, and frequency of physical activity The study will provide lifestyle guidance for healthy people and breast cancer patients, provide strategies to prevent breast cancer and improve prognosis, reduce the incidence and mortality of breast cancer, and reduce the medical burden of social funds.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-prospective-study-on-the-impact-of-clinical-characteristics-blood-proteomics-and-metabolomics-on-the-incidence-and-prognosis-of-urologic-cancers-in-the-uk-biobank

A Prospective Study on the Impact of Clinical Characteristics, Blood Proteomics, and Metabolomics on the Incidence and Prognosis of Urologic Cancers in the UK Biobank.

Last updated:
ID:
532881
Start date:
5 February 2025
Project status:
Current
Principal investigator:
Dr Zongren Wang
Lead institution:
The First Affiliated Hospital, Sun Yat-sen University, China

This project aims to investigate how clinical characteristics, blood proteomics, and metabolomics influence the incidence and prognosis of urological cancers (kidney, prostate, and bladder) in the UK Biobank cohort. We will address three key research questions: 1.Which clinical factors (e.g., age, BMI, lifestyle variables) are significantly associated with urological cancer incidence and survival? 2.How do proteomics and metabolomics markers improve risk stratification and prediction of disease outcomes? 3.Can machine learning models leveraging these multi-dimensional data offer clinically valuable diagnostic and prognostic tools?Objectives:Conduct Cox proportional hazards analyses to identify associations between baseline clinical features and cancer outcomes.Evaluate the predictive utility of selected protein and metabolic biomarkers using both traditional and machine learning approaches.Develop and validate diagnostic/prognostic models, comparing their accuracy to established clinical risk scores.Scientific Rationale:Although multiple studies have examined risk factors for urological cancers, few have integrated comprehensive proteomic and metabolomic data at a large scale. By leveraging the breadth and depth of UK Biobank data, our research will clarify the interplay between traditional risk factors and molecular biomarkers. The resulting findings may guide personalized risk assessment, improve early detection, and inform targeted prevention strategies for urological malignancies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-quantitative-covariate-adjustment-method-for-detecting-genome-wide-gene-gene-interactions-for-bladder-cancer-risk

A quantitative covariate adjustment method for detecting genome-wide gene-gene interactions for bladder cancer risk.

Last updated:
ID:
82999
Start date:
18 March 2022
Project status:
Closed
Principal investigator:
Miss Siting Li
Lead institution:
Dartmouth College, United States of America

This project will provide a general method for genome data analysis that can be potentially apply to any specific disease. We focus the application of this method on an investigation into the role of genetic polymorphisms in bladder cancer. This will advance our knowledge of bladder cancer and may help to improve diagnosis, prevention, and treatment for bladder cancer.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-replication-study-of-pain-interactions-with-comorbidities

A replication study of pain interactions with comorbidities

Last updated:
ID:
20802
Start date:
24 May 2016
Project status:
Current
Principal investigator:
Professor Luda Diatchenko
Lead institution:
McGill University, Canada

Full cohort, with the possibility of adding data from the future web-based questionnaire on pain.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-retrospective-cohort-analysis-regarding-the-association-of-childhood-trauma-experiences-with-bone-health-among-uk-biobank-participants

A retrospective cohort analysis regarding the association of childhood trauma experiences with bone health among UK biobank participants

Last updated:
ID:
143143
Start date:
18 January 2024
Project status:
Current
Principal investigator:
Mr Yangyang He
Lead institution:
University of Potsdam, Germany

Aims: Our research project aims to investigate the association between childhood trauma experience (measured by Childhood Trauma Screener) and bone health markers in adults, including bone-related serum markers and bone mineral density, through retrospective analyzing a large database cohort, thus to infer that whether childhood trauma experience has an association with bone health. We also aim to explore further which Childhood Trauma Screener subtype is associated with bone health.

Scientific rationale: Our previous research has found that the experience of childhood trauma impact bone health in adults, including reduced bone mineral density and dysregulated serum bone markers through animal and human studies. Now we try to take the research a step further, by analyzing the data from the UK Biobank database, we can explore the association between childhood trauma and bone health in a large cohort, updates our understanding of osteoporosis etiology, and open new avenues for personalized prevention and intervention strategies.

Project duration: The estimated duration of this project is 36 months.

Public health impact: The results of this study allow for additional beneficial outcomes for public health. By identifying potential associations with childhood trauma, the results of this study have the potential to provide early prevention strategies and increase awareness among healthcare practitioners, policymakers, and the public. Then will ultimately contribute to reducing the population’s childhood trauma burden.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-retrospective-investigation-of-accelerometer-derived-movement-patterns-with-the-aim-of-validating-a-machine-learning-algorithm-and-potential-identification-of-novel-signs-of-disease

A retrospective investigation of accelerometer-derived movement patterns, with the aim of validating a machine learning algorithm, and potential identification of novel signs of disease

Last updated:
ID:
44154
Start date:
24 September 2018
Project status:
Closed
Principal investigator:
Dr Claudiu Mihaila
Lead institution:
Lux Health Technologies Ltd, Great Britain

We aim to look at the movement data in UK Biobank data using a computer program that has been designed to spot patterns in large volumes of data. We will anonymously group people by medical history, age and gender (amongst other criteria), and observe for similarities within these groups. We will then tell the computer program what some of these represent, and it will learn to recognise changes from these patterns in the future – for example, the tremor observed in Parkinsonism and the limp seen in Osteoarthritis of the hip. It is possible that the program will discover similarities that have not been previously noticed by people. This might mean that we are able to discover new patterns of movement, that can be used to help diagnose people with illnesses in the future.

We believe that this project will take 1-2 years to complete fully, as the development of the computer program will be dependent on the data that we receive (and so it is very difficult to predict the duration).

This project presents the potential to help diagnose illness on a large scale by using the technological advancements (for instance, smart watches) that people already use. The opportunity to find new movement patterns is also an exciting one, and may be something that could help people even without access to wearable technology.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-retrospective-study-of-cardiovascular-event-predictions-using-integrative-and-interpretable-deep-learning-algorithms

A retrospective study of cardiovascular event predictions using integrative and interpretable deep learning algorithms

Last updated:
ID:
55051
Start date:
1 March 2021
Project status:
Current
Principal investigator:
Dr Bo Wang
Lead institution:
University Health Network, Canada

CVD is the second leading cause of death in Canada. Every year, approximately 70,000 Canadians die from a CVD-related illness and many more suffer life-threatening CVD events such as myocardial infarctions and strokes.
The research aims of the proposed study are:
Primary objective: to build, validate and optimize a multimodal architecture using deep learning algorithms to predict cardiovascular outcomes based on imaging data (MRI and ultrasound), ECG and patient clinical information.
Secondary objective: To determine new clinical prognostic parameters of cardiovascular outcomes based on the multimodal architecture, and to determine the impact of the population- and sex- differences in cardiovascular outcomes.
Most of the existing methods have been focusing on analyzing 2D images analysis rather than 3D images that are commonly prescribed to diagnose CVD. Moreover, most of the existing methods solely focus on analyzing images but neglect other information such as health records that can also be crucial in diagnosing CVD.
Our study aims to create a deep learning multimodal architecture to predict CVD outcomes by integrating complex imaging segmentation analysis for 3D images and multimodal data input such as clinical notes and ECG records. Our proposed architecture is promising to outperform the current standards given that deep learning methods are recognized to identify non-linear relationships in high-dimensional data.
The proposed study is expected to accelerate the process of 3D image analysis, to improve the accuracy of CVD diagnosis by incorporating 3D images analysis results with other clinical measurements, and ultimately to facilitate practitioners in better clinical decision-making.
The estimated duration of the study is 36 months: 6 months for data processing, 18 months for the primary objective and 12 months for the secondary objective.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-risk-benefit-analysis-for-the-use-of-serotonin-reuptake-inhibitors-ssris-as-a-first-line-pharmacological-treatment-for-major-depressive-disorder-in-frail-primary-care-patients

A risk-benefit analysis for the use of Serotonin Reuptake Inhibitors (SSRIs) as a first line pharmacological treatment for major depressive disorder in frail primary care patients.

Last updated:
ID:
300210
Start date:
27 November 2024
Project status:
Current
Principal investigator:
Ms Safoora Azimi-Yancheshmeh
Lead institution:
University of Brighton, Great Britain

Frailty is characterised by weakening of body’s normal functioning and can cause unwanted health problems such as hospitalisation, falls, sensitivity to unwanted effects of medications and death. Frailty can significantly increase the health-care costs and is a global health concern, and its incidence is expected to rise due to the rapid increase in ageing population.
Depression is one of the most common mental health illnesses worldwide and can have significant negative effect on the quality of life of patients and their care givers and cause death mainly due to suicide.
Depression is treated mostly in primary care settings such as GP surgeries in the UK. A group of medications called Selective Serotonin Reuptake Inhibitors (SSRIs) are the first line antidepressant drugs for treatment of depression in adults in the UK. They can cause unwanted effects such as falls, irregular heartbeat, anxiety, weakness of bones, nausea and feeling sick, bleeding, and sexual problems. They can also interact with other medications.
Frail patients generally have higher possibility of being diagnosed with depression. Frail patients may respond less favourably to the antidepressants and are more sensitive to their unwanted effects. Many frail patients have other illnesses and therefore are often on numerous medications which can interact with antidepressants and induce further risks and complications.
There is a significant gap of knowledge in relation to the role of SSRIs in causing or worsening frailty. The current treatment guidelines for treatment of depression do not provide sufficient advice about risks and benefits of using SSRIs for treatment of depression in frail patients.
This research investigates whether the use of SSRIs for treatment of depression can cause or worsen frailty in primary care patients. It also investigates the risks and benefits of using SSRIs as the first line antidepressants for treatment of depression in frail primary care patients and possible links between genetic and patient characteristics and responding to the SSRIs or having unwanted effects.
This project aims to develop a screening tool to help clinicians to prescribe SSRIs for frail patients when are expected to be effective and less likely to cause unwanted side effects to reduce the harm to patients and ensure effective treatment and is expected to last 36 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-risk-benefit-ratio-of-sunlight-exposure

A risk-benefit ratio of sunlight exposure

Last updated:
ID:
3932
Start date:
11 July 2013
Project status:
Closed
Principal investigator:
Professor Richard Beresford Weller
Lead institution:
University of Edinburgh, Great Britain

The prevalence of hypertension correlates with latitude, with BP rising with distance from the equator. Vitamin D is made in sun exposed skin, and serum levels correlate inversely with hypertension and ischaemic heart disease(IHD) prevalence. However, oral vitamin D supplements have no effect on BP or IHD. We hypothesise that sunlight has beneficial effects on cardiovascular disease and overall mortality, independently of vitamin D. We have demonstrated large stores of nitrogen oxides in the skin, released to the circulation on UVA irradiation of the skin, with a fall in BP.
Although sunlight exposure is risk factor for skin cancer there is no evidence that it causes a rise in all-cause mortality. epidemiological data suggest it may cause a reduction in deaths from hypertension related diseases, in particular cardiovascular and cerebrovascular disease. As deaths from these are almost 100 times more frequent than from skin cancer, the benefits of sunlight may exceed the risks. The UK Biobank dataset includes questions on sun exposure and latitude of residence and allow a prospective study on overall risk-benefit ratio of sunlight exposure. This meets the aspirations of the UK Biobank to improve prevention and treatment of disease, in particular cardiovascular and stroke.
We will seek access to the full dataset, looking at data on sunlight exposure and variables known to affect cardiovascular disease and stroke. Our primary outcome measure will be all-cause mortality and we will derive an odds-ratio for the effect of sunlight on this. We will also look for rates of skin cancer. We will correct for confounding factors that may link with different sun exposure patterns. We require access to data only (i.e. no samples) for the full cohort, with data on vitamin D serum levels when available


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-scale-for-your-liver-estimating-liver-fat-percentage-with-accessible-measurements

A Scale for Your Liver – Estimating Liver Fat Percentage with Accessible Measurements

Last updated:
ID:
50783
Start date:
15 January 2020
Project status:
Closed
Principal investigator:
Professor Sebastian Thrun
Lead institution:
Stanford University, United States of America

Livers have the second longest transplant waitlist in the US next to kidneys. Alcohol and obesity are known causes of fat build up in the liver. This fat build up leads to scarring. 1 in 4 people in the US and Europe have livers that are over 5% fat and experience no symptoms. The only good options for quantifying liver fat percentage are an invasive liver biopsy or an expensive MRI, both of which require significant suspicion of liver damage or other problems to justify. A tighter feedback loop would allow healthy minded individuals to correct deleterious behavior before reversible liver fat becomes irreversible liver scarring.

We aim to combine the latest advances in AI with routine blood and urine measurements to create a tool with which the healthy man and woman can better monitor their liver health. The current medically accepted fatty liver calculator uses 2 blood measurements in conjunction with body weight and height. It was trained on data collected from 496 people. The UK Biobank dataset contains data from over half a million people, and tons of blood and urine measurements. Our scientific rationale is that we can use this enormous dataset to train a more reliable fatty liver estimator.

When granted access to the dataset, we expect the project to be completed in under 3 months. If the trained estimator is reliable, we plan to release it to the public as an app. The public health impact of this work will be to allow better monitoring of liver health with data collected from routine blood and urine lab measurements.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-secondary-data-analysis-case-control-study-to-evaluate-dietary-intake-before-a-colorectal-cancer-diagnosis-in-the-british-population

A Secondary Data Analysis Case-control Study to Evaluate Dietary Intake before a Colorectal Cancer Diagnosis in the British Population.

Last updated:
ID:
41002
Start date:
14 May 2019
Project status:
Closed
Principal investigator:
Mrs Amal Abdullah Aldossari
Lead institution:
University of Manchester, Great Britain

This project aims to determine whether there is a difference between colorectal cancer patients one year pre-diagnosis and people without cancer diagnoses in their dietary intakes based on healthy eating recommendations. There is evidence highlighted that adopting a healthy diet may help in cancer prevention and improve survivorship. Furthermore, higher rates of mortality and morbidity noted in cancer survivors compared to people who have never had cancer, have motivated people living with and beyond cancer to change their lifestyle to a healthier one. Most studies assessed dietary intake before and after diagnosis among breast cancer patients, while very few studies evaluated the same for colorectal cancer patients. Therefore, the present study aims to evaluate dietary intake before and after diagnosis among colorectal cancer patients in the UK. The study’s findings might help patients understand how a healthier diet can facilitate cancer prevention and survivorship as diagnoses become more prevalent. In addition, the study will determine whether colorectal cancer patients tend to change their eating habits after diagnosis and whether this creates an opportunity for overall lifestyle and health improvement. This project will take 36 months from bio bank application submission until disseminate results.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-series-of-investigations-exploring-the-relationship-between-genetic-and-environmental-factors-and-acute-and-chronic-ocular-and-neurological-conditions

A series of investigations exploring the relationship between genetic and environmental factors and acute and chronic ocular and neurological conditions

Last updated:
ID:
76765
Start date:
5 November 2021
Project status:
Current
Principal investigator:
Professor Helen Victoria Danesh-Meyer
Lead institution:
University of Auckland, New Zealand

Aims: Our overarching aim is to explore eye-brain function relationships. Glaucoma is a condition which there is gradual damage to the optic nerve, or the nerve of sight. While the optic nerve is located at the back of the eye it is actually an extension of the brain or central nervous system. It is recognised that one of the processes that contributes to glaucoma progression is neuroinflammation, or inflammation of the nervous system. The focus of this research is to identify and evaluate biological, behavioural, and environmental factors in the aetiology of glaucoma and related neurodegenerative disorders in which neuroinflammation plays a significant role.

Scientific Rationale: Glaucoma is regarded as a neurodegenerative disorder. However, the evidence for this hypothesis is controversial, as it is based on generally smaller studies that have reported variable results. Using the powerful and detailed database of UK Biobank, we will robustly evaluate multiple biological, behavioural, and environmental factors implicated in neurodegenerative pathways to determine their causal relevance to glaucoma and related neurodegenerative conditions, thereby identifying modifiable risk factors for glaucoma. In addition, we will develop risk scores which incorporate environmental, biological, physical and genetic variables, thereby advancing risk prediction of glaucoma.

Project duration: 3 years

Potential Public Health Impact: Glaucoma is a leading cause of blindness. Presently, there are no significant modifiable risk factors other than intraocular pressure. This investigation will identify modifiable environmental, physical, biological and genetic biomarkers in relationship to glaucoma. Furthermore, it will explore the association between glaucoma and neurodegenerative pathways which will allow a deeper understanding of the underlying mechanisms that impact the disease process


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-social-ecological-model-of-subjective-well-being-versus-ill-being-a-structural-equation-modelling-study-on-the-uk-biobank-cohort

A social ecological model of subjective ‘well-being’ versus ‘ill-being’: A structural equation modelling study on the UK Biobank cohort

Last updated:
ID:
92623
Start date:
26 October 2022
Project status:
Closed
Principal investigator:
Dr Darren Edwards
Lead institution:
Swansea University, Great Britain

Aims,
This project aims to improve our understanding of the pathways to subjective wellbeing versus ill-being, taking into account key variables at multiple levels of scale, spanning the individual, community and environment.
Scientific rationale,
Prior research has identified a number of key variables influencing human wellbeing, ranging in scale from biopsychsocial variables (e.g. the beat-to-beat variability in heart rate, cognitive flexibility and social connectedness) to variables relating to sociocontextual and ecological impacts (e.g. social capital, and green space availability etc). Research has seldom accounted for such complexities and potential interactions stemming from the interconnectedness of multiple systemic factors influencing the complex construct of wellbeing and illbeing. This work will therefore conduct structural equation modelling and path analysis to explore more sophisticated model of wellbeing and examine its relevance to related yet distinct construct of illbeing, taking into account the many mediating and moderating factors of key wellbeing markers.
Project duration,
2 Years.
Public health impact,
We will develop a contemporary evidence-based model that reflects the complex construct of individual wellbeing. By incorporating ecological, social, psychological, and physiological influencing factors, this work will facilitate a transdisciplinary approach to studying public health relevant factors of ‘wellbeing’ versus ‘ill-being’. In doing so, we hope to aid a conceptual shift within wellbeing research that will open up avenues for interventions promoting positive change at individual, social, and ecological levels.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-statistical-framework-for-personalized-nutrition-recommendations-based-on-genetic-and-anthropometric-data

A statistical framework for personalized nutrition recommendations based on genetic and anthropometric data.

Last updated:
ID:
28784
Start date:
1 July 2017
Project status:
Current
Principal investigator:
Professor Eran Segal
Lead institution:
Weizmann Institute of Science, Israel

In recent years there is accumulating evidence that genetics and lifestyle habits interplay in complex ways to affect clinical conditions. We intend to investigate the relationship between genetic factors, dietary habits, anthropometric traits and several clinical outcomes, with the aim of proposing personalized nutrition recommendations. Our main goal is to minimize the risk of obesity and other diet-induced disease via individual-specific nutrition recommendations. The proposed research aims to improve the prevention, diagnosis and treatment of illness and the promotion of health by providing personalized nutrition recommendations, in alignment with the UK biobank?s stated purpose. We intend to identify genetic mechanisms that, in combination with the dietary habits of an individual, can be used to predict clinical outcomes. These findings can be used to provide personalized nutrition recommendations to minimize risk for adverse outcomes. We intend to use all individuals for which both genetic and dietary habits data is available to obtain as much power as possible to achieve significant results (approximately 500,000 individuals).


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-statistical-framework-to-discover-sex-specific-genetic-effect

A Statistical Framework to Discover Sex-Specific Genetic Effect

Last updated:
ID:
46030
Start date:
13 February 2019
Project status:
Current
Principal investigator:
Dr Nikolaos Patsopoulos
Lead institution:
Brigham and Womens Hospital, United States of America

Our primary aim is to discover the sex-specific genetic variants, regardless of the way they are associated with a certain disease or trait. By setting this aim, we seek to answer the question that 1) what are the genetic variants that have sex-specific effects on a certain disease or traits. And 2), are those variants impacting the disease or traits through their additive effect, other genetic models, or their interactions with sex.

Many diseases, including most of the autoimmune related diseases, show strong disproportionality between genders. Previous researches often focus either the effect of the variant itself, or on interaction of variant with sex. However, there is no method to jointly estimate both effects and discover the sex-specific genetic architecture therefrom.

To achieve the above aim, we will propose a framework to 1) identify sex-specific genetic variants through statistical tests on the joint effect of additive model of the genetic variants, and their interaction with sex; and 2), further distinguish and categorize the types of effecting patterns of the variants, in order to further unravel the biological pathways and mechanisms involving those variants.

By addressing the above questions, discovering the sex-specific genetic architectures of the traits/diseases that shows strong variance in incident rates between sexes, we hope to shed light on finding the genetic causations on those diseases. And furthermore, by categorizing them by patterns of their sex-specific effects on the traits/diseases, we seek to deepen the understanding of the pathways and mechanisms of those diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-step-forward-in-understanding-the-complex-relationship-between-exposure-to-multiple-health-risk-factors-and-multiple-morbidity-a-study-based-on-the-uk-biobank-cohort

A step forward in understanding the complex relationship between exposure to multiple health risk factors and multiple morbidity: a study based on the UK Biobank Cohort

Last updated:
ID:
104055
Start date:
22 August 2023
Project status:
Current
Principal investigator:
Professor Carlo La Vecchia
Lead institution:
University of Milan, Italy

We are living longer. Consequently, with age we are likely to accumulate multiple chronic diseases like high blood pressure, Type 2 diabetes and heart diseases at the same time. Scientifically, this is referred as multimorbidity. At the moment if affects 27% of all adults in the UK however this is set to increase significantly in the future. This is going to be one of the biggest challenges not only National Health Service but for society as a whole.
We know that as we get older our health declines, however we also know that several modifiable risk factors such as lifestyle factors such as smoking, diet, alcohol intake, how much we move as well as things like pollution also affect our health. Time and time science has shown that people who move less, smoke or have an excessive intake of alcohol and who eat an unhealthy diet are at a higher risk of developing several chronic diseases like heart disease and cancer. New research shows that exposure to unhealthy levels of air pollution adds to the risk of these chronic diseases. Other research also shows that there are non-medical factors such as the level of education, social class, income and the neighbourhood you live in also play their role in our health. However, there is little research that has looked at the impact of all these factors on health.
Each of these factors (sociodemographic, psychosocial, lifestyle and environmental factor) all play a role in the development of multiple chronic conditions. However, what we don’t yet know is how these risk factors which coexist within the same individual interact together. Studying this specifically is a challenge and as a result standard statistical techniques are of limited use in this regard.

Using the UK Biobank Cohort we aim to try understand the complex relationship between the exposure to multiple health risks and the development of multiple chronic diseases. The expected results of this study will help to identify which groups may be more at risk of developing multiple diseases in the future and will require much more targeted public intervention.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-study-integrating-demographic-clinical-data-genomic-and-proteomic-mendelian-randomization-and-machine-learning-model-development-in-patients-with-hemorrhagic-transformation-of-ischemic-stroke

A study integrating demographic, clinical data, genomic and proteomic Mendelian randomization and machine learning model development in patients with hemorrhagic transformation of ischemic stroke

Last updated:
ID:
946047
Start date:
15 August 2025
Project status:
Current
Principal investigator:
Dr Xiaoming Ma
Lead institution:
Suzhou Hospital, Affiliate Hospital of the Medical School of Nanjing University, China

We propose to identify causal protein and genetic determinants of hemorrhagic transformation (HT) in ischemic stroke patients through two-sample or Bi-directional Mendelian randomization (MR) using plasma pQTLs and large!scale GWAS summary statistics from MEGASTROKE and UK Biobank. We aim to develop and validate a high performance machine-learning (ML) based risk prediction model for HT by integrating demographic, clinical, genomic, and proteomic features using algorithms like XGBoost. We will interpret model predictions with SHapley Additive exPlanations (SHAP) to elucidate each feature’s contribution and nonlinear relationships with HT risk. Our objectives include assessing discrimination (AUC) and calibration (calibration curves, decision curve analysis) through nested cross!validation and external validation cohorts. We will perform colocalization analyses to confirm shared causal signals between pQTLs and stroke outcomes. Model hyperparameters will be optimized via grid search to balance performance and generalizability.
Finally, we will generate SHAP-derived risk scores and share all analytic code and derived variables within six months of publication to enhance UK Biobank resource utility.
Last, we need samples data to identify potential biomarkers(including gene, proteins, clinicasl data and samples) for predict HT in early stage by using ML methods.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-study-investigating-leukocyte-counts-in-women-with-self-reported-polycystic-ovarian-syndrome-results-from-the-uk-biobank-prospective-study

A Study Investigating Leukocyte Counts in Women with Self-reported Polycystic Ovarian Syndrome: Results from the UK BioBank Prospective Study.

Last updated:
ID:
46654
Start date:
20 June 2019
Project status:
Closed
Principal investigator:
Dr Blake John Cochran
Lead institution:
University of New South Wales, Australia

Polycystic Ovarian Syndrome (PCOS) is a common hormonal condition affecting approximately 5-20% of women worldwide. The condition results in the ovaries producing excessive male hormones. This hormonal imbalance can result in irregular or absent menstrual periods, difficulty becoming pregnant and mood disturbances. The condition also increases the chance of developing cardiovascular disease and diabetes even after menopause.

The exact cause of PCOS and how it increases the chance of developing cardiovascular disease and diabetes is not known. Research shows that there may be a genetic link and that PCOS can run in families. There also appears to be a connection with obesity, although the details of how this occurs is not clear.

Recent research has identified long-term inflammation as a possible link between PCOS and insulin resistance. Insulin is a hormone that allows cells in the body to take up glucose, so women with PCOS often develop a state in which their cells cannot use insulin. As a result, the pancreas can produce more insulin to make up for this and over time can lead to diabetes. Women with PCOS also develop abnormalities and inflammation in their fat tissue, which is thought to contribute to diabetes and cardiovascular risk.

Inflammation often results in involvement of the body’s immune system, which include white blood cells. There is not enough current information about how the inflammation that occurs in PCOS affects white blood cell numbers. This study therefore aims to answer this question by using the UK BioBank data which includes 273,469 women, of whom 643 have self-reported as having PCOS. The project duration is expected to be three to six months, but may be longer as more data becomes available. Answering this research question has the potential to better understand PCOS and how it increases the risk of cardiovascular disease and diabetes. With this greater understanding, novel treatment options can be developed to reduce the morbidity associated with PCOS.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-study-of-decision-making-using-uk-biobank-and-fmri-studies-of-economic-choices

A study of decision making using UK Biobank and fMRI studies of economic choices

Last updated:
ID:
31028
Start date:
15 March 2018
Project status:
Current
Principal investigator:
Professor Aldo Rustichini
Lead institution:
University of Minnesota Twin Cities, United States of America

The main goal of the research is the systematic integration of genetic and brain activation data to explain the underpinnings of economic and social choice, and use the results to understand neurocognitive and mental health outcomes. We develop a novel approach integrating functional neuro-imaging and the genetic data using both Bayesian and classical statistical methods.

We plan to build a Hierarchical Bayesian model and use other classical statistic method of decision making. Analysis of UK Biobank data will be cross-validated with independent samples available at the University of Minnesota, relevant for the study of schizophrenia and addiction. The research will address health-related questions in the public interest both directly and indirectly.

Directly, through the application of the study to causal factors of schizophrenia and addiction (both drug/alcohol and behavioral).

Indirectly because it will improve the understanding of the impact of cognitive functions on important life outcomes, which eventually affect health conditions. The specific aim of this study is to examine association signals from SNPs, examining how much of the variation is captured by examining all SNPs simultaneously (using Polygenic Risk Scores), and look at the extent to which SNPs that predict variation in two joint sets of phenotypes (in our case, patterns of brain activation and choice).

For this purpose we will use statistical techniques (Gene Set Enrichment Analysis, Group Lasso) testing the hypotheses that specific biological pathways affect jointly the choices and brain activity implementing it. We will integrate several data sets for cross-validation. The research requires data on the full cohort of subjects available. In particular we need the full genotype data and the raw imaging data. We are aware that this will require large data storage, so a clarification is needed. In our team the effort is aimed at integrating the analysis of the UK Biobank data and the Human Connectome Project (http://www.humanconnectome.org/). For this we need in addition to functional and T1 data the T2 data. We also like to be sure that the geometric B0 correction is done properly.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-study-of-genes-implicated-in-leber-congenital-amaurosis-lca-and-the-role-of-preconception-genetic-screening

A study of genes implicated in Leber congenital amaurosis (LCA) and the role of preconception genetic screening

Last updated:
ID:
44316
Start date:
29 May 2019
Project status:
Closed
Principal investigator:
Dr Michael A Grassi
Lead institution:
University of Illinois at Chicago, United States of America

Children with LCA are born blind.  Even though the disease is rare, it has significant long term healthcare implications. A number of different genes have been implicated in the pathogenesis of LCA. Alleles are different forms of the same gene located on the same place on the chromosome. Comparing the frequency of different alleles in the causation of LCA will give a fair idea as to the mutations responsible. This will help in better screening techniques that can be used in the population, to reduce the incidence and prevalence of the disease, thus resulting in reduced blindness in the overall population. Reduced cases of LCA will mean easing the dependency on the healthcare system resources, even if by very little.
We expect the project to last less than 6months, since it will be done insilico, with a simulated computer program.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-study-of-genetic-factors-predisposing-for-critical-illness

A study of genetic factors predisposing for critical illness

Last updated:
ID:
91813
Start date:
10 August 2022
Project status:
Current
Principal investigator:
Professor Miklos Lipcsey
Lead institution:
Uppsala University, Sweden

Critical illness leading to intensive care means suffering and high risk of death for the individual, and costs to society. There are several reasons why patients end up in intensive care units (IVA), but common to them is that they develop failure in vital organs. Sepsis, COVID-19 infection, and surgical trauma are common causes of failure of vital organs. During the past year, these conditions have accounted for a very large proportion of all admissions to ICU in Sweden.
We have previously shown that inherited a.k.a. genetic factors are important in how ill a person gets from the same infection or surgical procedure. The planned study can help us understand which genetic factors are important risk factors for getting severely ill and recovering thereafter. Moreover, these studies can help us understand some of the mechanisms leading to severe illness, increasing the chance of finding treatments to prevent or cure these conditions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-study-of-influencing-factors-for-cerebrovascular-and-neurodegenerative-diseases

A Study of influencing factors for cerebrovascular and neurodegenerative diseases

Last updated:
ID:
104830
Start date:
11 July 2023
Project status:
Current
Principal investigator:
Professor Jun Lyu
Lead institution:
Jinan University, China

Aims:
Our aim is to explore the association between influencing factors (including biochemical indices, plasma metabolites, lifestyle, environmental factors and genetic factors) and the incidence and progression of cerebrovascular and neurodegenerative diseases, and investigate the association of influencing factors and the images of brain diseases.

Scientific rationale:
Cerebrovascular and neurodegenerative diseases are important public health issues worldwide, causing serious morbidity, mortality and economic burden. Despite extensive previous research efforts, the influencing factors of these diseases remain incompletely understood. We propose a study to identify modifiable and non-modifiable influencing factors for cerebrovascular and neurodegenerative disease using the UK Biobank data. By identifying these factors, we hope to improve our understanding of the pathogenesis of cerebrovascular and neurodegenerative disease, and contribute to the development of more effective prevention and treatment strategies.

Project duration:
This project is expected to last for 36 months.

Public health impact:
By identifying key contributing factors, public health policies can be developed to help mitigate the incidence, ultimately improving population health outcomes and reducing the economic burden associated with cerebrovascular and neurodegenerative diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-study-of-risk-factors-and-protective-factors-for-neuroimmune-disorders

A Study of risk factors and protective factors for neuroimmune disorders

Last updated:
ID:
442119
Start date:
25 June 2025
Project status:
Current
Principal investigator:
Ms Wanning Li
Lead institution:
Huazhong University of Science and Technology, China

Neuroimmune diseases, including multiple sclerosis (MS), neuromyelitis optica spectrum disorders (NMOSD), anti-myelin oligodendrocyte glycoprotein-IgG (MOGAD), Guillain-Barre syndrome (GBS) etc., predominantly occur in young and middle-aged individuals with a high rate of disability. The pathogenesis of these diseases remains unclear, and the treatment outcomes are often unsatisfactory. Therefore, exploring the prevention and treatment of neuroimmune diseases is of vital importance. In this study, we will identify risk factors and protective factors influencing disease onset and prognosis from multiple perspectives based on large-sample population data.
In our research, first we will collect baseline data from the population (including general demographic characteristics, medical history, and laboratory data) as well as outcome events (the onset and prognosis of autoimmune diseases of the nervous system). The population will be grouped, and various statistical methods (such as logistic regression, Cox regression and so on) will be employed to compare the differences in various factors between groups, with the aim of identifying potential protective and risk factors that influence the onset and prognosis of neuroimmune diseases. We also intend to compare factors related to neuroimmune disorders with neurodegenerative diseases, as well as anxiety, depression, and other psychosomatic disorders, which will be more conducive to identifying the signature indicators of neuroimmune that we are concerned about and to finding common factors that affect neurological diseases. Furthermore, we will explore the relationship between various factors and autoimmune diseases of the nervous system at the genetic level.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-study-of-risk-factors-for-cardiovascular-and-cerebrovascular-diseases

A Study of risk factors for cardiovascular and cerebrovascular diseases

Last updated:
ID:
94533
Start date:
10 November 2022
Project status:
Current
Principal investigator:
Miss Lian Chen
Lead institution:
Huazhong University of Science and Technology, China

Cardiovascular and cerebrovascular diseases are the primary diseases that threaten human life and health. At present, some related risk factors have been identified and treatment methods have been applied. However, the prognosis is still poor. Therefore, the purpose of our study is to find the risk factors of diseases from multiple perspectives based on the data of a large sample population, including general demographic characteristics, disease history, medication history, blood biochemical indexes and so on. On the basis of traditional risk factors, we can find more connections, such as the association among cardiovascular and cerebrovascular diseases. Therefore, this study aims to guide the precise management of cardiovascular and cerebrovascular diseases, find new risk factors, explore new targets for prevention and treatment, improve prognosis, reduce disease burden, and contribute to human health.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-study-of-the-impact-of-physical-exercise-and-activity-level-on-cognitive-function-and-associated-mri-brain-changes-in-chronic-kidney-disease

A study of the impact of physical exercise and activity level on cognitive function, and associated MRI brain changes, in chronic kidney disease.

Last updated:
ID:
63941
Start date:
8 November 2021
Project status:
Current
Principal investigator:
Professor Indranil Dasgupta
Lead institution:
University Hospitals Birmingham NHS Trust, Great Britain

Cognitive impairment (reduced brain functioning) is more common in people with kidney disease than in those without it. We know that people who have cognitive impairment do not engage well with the healthcare system, have a poorer quality of life and are more likely to die early.

Being physically active is known to improve brain function in the general population. Research has shown some benefits of physical exercise in people with severe kidney problems who are on dialysis treatment. However, this has not been studied in detail in those with less severe forms of kidney disease. By studying the effects of physical activity on brain function in people with kidney disease, we may find a new treatment to help to prevent and slow down cognitive impairment in those with kidney disease.

In this study we aim to;

1. Find out how common cognitive impairment is in those with kidney disease in the UK Biobank study population.
2. Find out if there is a relationship between physical activity level and cognitive function in those with kidney disease.
3. Find out if people with lower physical activity levels and kidney problems are more likely to develop cognitive impairment and die than those who have higher activity levels.
4. Find out if we can see where and what type of damage in the brain occurs in these people, by looking at MRI brain scans, and assess relationship with physical activity. This may also help us to better understand what processes are involved in this damage.
5. Find out what other factors might be involved, such as risk factors for heart disease and inflammation (body’s response to infection or injury), in cognitive impairment in people with kidney disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-study-on-genetic-variations-associated-with-dizziness-using-the-uk-biobank-data

A study on genetic variations associated with dizziness using the UK Biobank data.

Last updated:
ID:
104323
Start date:
27 June 2023
Project status:
Current
Principal investigator:
Professor Byung Yoon Choi
Lead institution:
Seoul National University Bundang Hospital, Korea (South)

Dizziness is a common complaint among patients who visit primary care physicians, neurologists, and otolaryngologists. Some types of dizziness have been found to be familial, which distinguishes them from other causes of dizziness. Familial dizziness has been observed in patients with isolated recurrent attacks of dizziness, hereditary deafness syndromes, and in patients with neurological disorders. While research in these areas has advanced our understanding of dizziness, much remains to be elucidated about its underlying causes. Through analyzing the genome data of the UK Biobank, we aim to identify genetic mutations that may contribute to development of diseases that can cause dizziness. By identifying genetic factors associated with dizziness, researchers can gain insights into the underlying mechanisms of the disorder and potentially develop more targeted and effective treatments. Additionally, identifying individuals at a higher risk for certain types of dizziness would allow earlier intervention and prevention based on their genetic profile.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-study-on-risk-factors-and-genetic-susceptibility-of-skin-diseases-and-comorbidities-or-cancers-based-on-uk-biobank-data

A Study on Risk Factors and Genetic Susceptibility of Skin Diseases and comorbidities or Cancers Based on UK Biobank Data

Last updated:
ID:
843724
Start date:
16 July 2025
Project status:
Current
Principal investigator:
Miss Wenchao Li
Lead institution:
Hospital for Skin Diseases, Shandong First Medical University, China

Skin diseases are highly prevalent, often chronic, and frequently co-occur with systemic conditions such as cardiovascular disease, inflammatory bowel disease, and various malignancies. Approximately 30% of patients with skin disorders present at least one comorbidity, and their risk of cancer-related comorbidities is estimated to be two to five times higher than the general population. However, the underlying mechanisms remain poorly understood. This study aims to address three key questions: (1) What modifiable and genetic factors are associated with the onset of skin diseases and related comorbidities? (2) How do genetic predispositions interact with environmental triggers in influencing disease progression? (3) What shared biological mechanisms underlie these comorbidities?

To answer these questions, we will leverage the UK Biobank’s large-scale, multidimensional dataset-encompassing genetic, environmental, lifestyle, and clinical information-to identify key risk factors and infer causal relationships. We will apply advanced epidemiological and statistical methods, including Cox regression, GWAS, Mendelian randomization, and machine learning, to explore associations, interactions, and disease trajectories. This integrated approach will help uncover the complex interplay between genes and environment in skin disease etiology and comorbidity development. The findings are expected to provide a robust scientific foundation for early prevention, precise risk stratification, and the development of personalized intervention strategies, thereby improving patient outcomes and supporting public health efforts.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-study-on-the-impact-of-sleep-patterns-psychological-factors-environmental-exposures-lifestyle-and-clock-gene-polymorphisms-on-the-development-of-polycystic-ovary-syndrome-related-infertility-and

A Study on the Impact of Sleep Patterns, Psychological Factors, Environmental Exposures, Lifestyle, and Clock Gene Polymorphisms on the Development of Polycystic Ovary Syndrome-Related Infertility and

Last updated:
ID:
632861
Start date:
29 June 2025
Project status:
Current
Principal investigator:
Miss Ruyu Luo
Lead institution:
Huazhong University of Science and Technology, China

Existing studies show that women with PCOS have a significantly higher prevalence of sleep disorders compared to healthy controls, including obstructive sleep apnea, hypersomnia, and insomnia. Subclinical sleep issues such as poor sleep quality, difficulty falling asleep, fatigue, and nighttime awakenings are also common in PCOS. These sleep problems are linked to metabolic abnormalities like insulin resistance, hyperandrogenism, and obesity. However, the causal relationship between sleep disorders and PCOS onset remains unclear, and longitudinal studies on the impact of sleep disorders on PCOS-related comorbidities are lacking.
This study aims to fill this gap through longitudinal and mediation analyses, over a 36-month period. The primary goals are to explore the interactions between sleep patterns, psychological factors, clock gene polymorphisms, environmental exposures, and PCOS-related reproductive, metabolic, and psychological phenotypes. We will also investigate the causal relationship between sleep patterns and PCOS onset and the effects of sleep disorders on comorbidities. Findings will provide evidence for early prevention and personalized interventions for PCOS.
The UK Biobank cohort includes individuals aged 40-69 (2006-2010), many of whom are postmenopausal. Our research will focus on individuals diagnosed with PCOS before menopause, using clinical data (reproductive history, hormone levels) to identify potential PCOS cases. We will examine long-term health outcomes, such as metabolic and cardiovascular health, using longitudinal UK Biobank data, and have reviewed relevant research to identify suitable cases for the study.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-study-to-investigate-the-risk-factors-for-developing-ductal-carcinoma-in-situ-dcis-in-women-from-the-uk-biobank

A study to investigate the risk factors for developing Ductal Carcinoma in Situ (DCIS) in women from the UK Biobank

Last updated:
ID:
11091
Start date:
1 April 2015
Project status:
Current
Principal investigator:
Dr Gurdeep Mannu
Lead institution:
University of Oxford, Great Britain

Although DCIS is considered a pre-invasive condition, it can progress to invasive breast cancer or death over a number of years. It is unclear which women with DCIS will develop invasive cancer within their lifetime and over what period of time. This project will investigate the risk factors for developing DCIS and its subsequent progression to invasive cancer and aim to quantify a woman?s risk of disease progression based on her lifestyle, dietary, socioeconomic, medical and genetic factors in order to help clinicians tailor diagnosis/screening, follow-up and treatment options according to individual patient risk-profiles. The objective of the UK Biobank is to undertake research in improving the prevention, diagnosis and treatment of a wide range of serious illnesses including breast cancer. By investigating the risk factors for developing DCIS or having a poorer outcome after DCIS treatment, it may be possible to quantify a woman?s risk based on her individual risk factors and so tailor prevention, diagnosis, and treatment options accordingly. Hence the aim of this project is in synergy with that of the UK Biobank and could help to provide innovative new directions to improve healthcare for DCIS patients. Of the women within the UK Biobank diagnosed with DCIS, analyses will be conducted of patient, medical, socioeconomic, dietary, lifestyle and genetic factors to investigate possible risk factors for DCIS when compared against age matched controls without DCIS or invasive breast cancer and with those women who were diagnosed with invasive breast cancer without a prior diagnosis of DCIS. Further analyses of the women with DCIS will be conducted according to whether or not they were subsequently diagnosed with invasive breast cancer. Three groups ((i) DCIS patients (n=2000 women), (ii) age matched controls (n=8000 women), and (iii) primary invasive breast cancer patients (n=2000), ensuring at least 4 women as controls for each woman with DCIS or invasive breast cancer) will be included in this study with a total of approximately 12,000 women. After 12-18 months a further data request will be made in order to increase the sample size of the cohort further by including new incident DCIS cases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-subgroup-analysis-exploring-the-association-between-chronic-musculoskeletal-pain-constipation-and-colorectal-cancer-in-uk-population

A subgroup analysis exploring the association between chronic musculoskeletal pain, constipation and colorectal cancer in UK population

Last updated:
ID:
753719
Start date:
3 July 2025
Project status:
Current
Principal investigator:
Mr Edwin Lam
Lead institution:
The Hong Kong Polytechnic University, Hong Kong

Chronic musculoskeletal pain (CMP) is prevalent health issues that impact the quality of life of many individuals. CMP is defined as persistent or recurrent musculoskeletal pain deriving from musculoskeletal structures such as joint, muscles and bones and last for at least three months(Zhuang et al., 2022). It is commonly found among older adults, with a point prevalence of more than 50% among elderly (Yamada et al., 2022).

Constipation is also commonly occurs among patients with CMP. While it is known that constipation may lead to chronic back pain, evidence also suggests that constipation may affect gut microbiome, which in turn can affect pain perception (Arai et al., 2018; Smith et al., 2008).However, the current understanding on the association between CMP and constipation remains unclear.

In addition, constipation is frequently associated with colorectal cancer (CRC). In 2018, CRC was the fourth mostly commonly diagnosed cancer in the world, with approximately 2 million new cases (Rawla et al., 2019). However, for the severity of constipation or bowel movement frequency that could induce CRC, it remains unclear. By understanding the correlation between CMP, constipation and CRC, it could contribute to the development of comprehensive treatment strategies to improve the health-related quality of life (HRQoL) of these individuals.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-systematic-analysis-of-trans-ethnic-portability-and-within-family-validation-of-polygenic-risk-score-prediction

A systematic analysis of trans-ethnic portability and within-family validation of polygenic risk score prediction

Last updated:
ID:
80545
Start date:
8 November 2021
Project status:
Current
Principal investigator:
Dr Yuntao Xia
Lead institution:
Orchid Bioscience, Inc, United States of America

Over the last ten years, polygenic risk scores have advanced from academic subject matter to clinically useful tools, capable of assisting diagnoses and setting screening standards. Yet, as medical professionals embrace the use of these tools in the US healthcare system, researchers have warned that these predictions are worse than expected for non-Europeans and cannot stratify risk within families as well as expected. Over the last several years, statistical geneticists have developed theory for and empirically analyzed the causes of these differences in prediction accuracy by ancestry.

Prediction accuracy tends to get worse as the genetic distance between training cohort and test cohort increases, and this can even occur when applying polygenic risk score models trained on a British cohort to an Italian test cohort. To help fix these problems, researchers have developed a number of tools that help construct risk prediction models that take ancestry into account.

Within the next 36 months, we aim to systematically assess these methods across a variety of traits to compare their utility risk prediction in non-European cohorts. We further intend to investigate whether these gaps differ by the trait or disease analyzed.

The proposed research fills an important gap in the current literature because there is no standardized baseline. Since differences in risk prediction by ancestry create a barrier to equitable use in the clinic, this research addresses an important need in public health.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-systematic-investigation-of-biomarkers-and-risk-factors-associated-with-the-progression-of-cardiovascular-kidney-metabolic-syndrome

A systematic investigation of biomarkers and risk factors associated with the progression of Cardiovascular-Kidney-Metabolic Syndrome.

Last updated:
ID:
673112
Start date:
7 April 2025
Project status:
Current
Principal investigator:
Dr Aoran Huang
Lead institution:
The Eighth Affiliated Hospital of Sun Yat-sen University, China

Cardiovascular-kidney-metabolic syndrome (CKM) is a novel concept introduced by the American Heart Association (AHA) in 2023, which refers to a systemic disease resulting from the interactions between metabolic risk factors, chronic kidney disease, and the cardiovascular system. This syndrome can lead to multi-organ dysfunction and increase the risk of cardiovascular adverse events. However, the early diagnostic biomarkers for CKM have yet to be established, and the social and environmental factors involved in its progression remain unclear.

Objective
To address this gap, the present study aims to adopt an integrated approach that leverages extensive genetic, metabolomic, proteomic, and comprehensive epidemiological data. Through multidimensional analyses, this study seeks to identify novel risk factors and uncover potential biomarkers for CKM.

Scientific rationale
Probing circulating proteins provides unique opportunities to uncover novel biomarkers and improve our understanding of etiology of those diseases. Similarly, blood metabolome is considered as important readouts of aggregated information from genetic factors, gene expression to protein abundance, as well as external environmental factors. To accomplish this, we will combine multi-omics (i.e., proteomics and metabolomics) and epidemiological data to discover novel risk factors, biomarkers and provide definitive evidence for known CKM risk factors reported by traditional observational studies. Our endeavor will encompass the analysis of multi-omics and epidemiological information, enabling us to unravel new risk factors, identify potential biomarkers, and comprehend the intricate web of causal relationships underpinning CKM. This comprehensive exploration will encompass conditions such as diabetes, chronic kidney disease, atherosclerosis, vascular calcification, and others.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-systems-biology-approach-to-neurological-disorders-integrative-analysis-of-multi-organ-imaging-multi-omics-lifestyle-and-environmental-exposures

A Systems Biology Approach to Neurological Disorders: Integrative Analysis of Multi-Organ Imaging, Multi-Omics, Lifestyle, and Environmental Exposures

Last updated:
ID:
854614
Start date:
17 September 2025
Project status:
Current
Principal investigator:
Dr Jingxin Yan
Lead institution:
South China University of Technology (SCUT), China

Neurological disorders such as Alzheimer’s disease, Parkinson’s disease, and stroke result from complex interactions among genetic, systemic, behavioral, and environmental factors. This project aims to explore how multi-organ imaging phenotypes, multi-omics data (genomics, proteomics, metabolomics, epigenomics), lifestyle behaviors, and environmental exposures contribute to the risk and development of neurological disorders.
We will address the following key questions:
How do brain and peripheral organ structural changes relate to neurological outcomes?
Which circulating biomarkers and molecular profiles are predictive of disease onset?
How do genetic and epigenetic factors interact with modifiable exposures such as air pollution, diet, and physical activity?
Can integrative models combining imaging, omics, and environmental data improve early disease prediction?
This project seeks to develop a systems-level understanding of neurological disorders, with potential applications in risk stratification, biomarker discovery, and targeted prevention strategies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-three-way-bi-directional-mendelian-randomization-analysis-of-diabetes-chronic-kidney-disease-and-coronary-heart-disease

A three-way bi-directional Mendelian randomization analysis of diabetes, chronic kidney disease and coronary heart disease

Last updated:
ID:
24117
Start date:
1 November 2016
Project status:
Closed
Principal investigator:
Professor Deborah Lawlor
Lead institution:
University of Bristol, Great Britain

Traditional observational studies have reported positive associations among (a) type 2 diabetes, (b) chronic kidney function, and (c) coronary heart disease. However, whether the association is due to confounding or reverse causality is unclear. This proposal aims to investigate the causal association among these 3 health conditions with each other using Mendelian randomization instrumental variable analysis. The proposed research will help investigate how these diseases affect each other using Mendelian randomization design which is less susceptible to confounding than observational studies. The results will provide additional insights concerning the bi-directional association among 3 major health threats (cardiovascular disease, diabetes and renal disease) with corresponding implications for etiology and therapeutics. We compare risk of cardiovascular disease, type 2 diabetes and renal disease, according to genetically determined per unit of risk difference (per odds of) for these three diseases. 500,000 participants (full cohort) for the analysis involving cardiovascular disease, type 2 diabetes, renal disease and genetic data.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-two-stage-polygenic-risk-score-and-mendelian-randomization-study-to-decipher-the-shared-genetic-architecture-between-cerebrovascular-disease-and-alzheimers-dementia

A Two-Stage Polygenic Risk Score and Mendelian Randomization Study to Decipher the Shared Genetic Architecture between Cerebrovascular Disease and Alzheimer’s Dementia.

Last updated:
ID:
1024568
Start date:
12 October 2025
Project status:
Current
Principal investigator:
Ms Zheng Tu
Lead institution:
Wenzhou People's Hospital, China

Global dementia cases are projected to exceed 80 million by 2040, with Alzheimer’s disease (AD) and cerebrovascular contributions representing major etiologies. Despite clinical co-occurrence, the genetic interplay and causal pathways between cerebrovascular disease (CeVD) and AD remain poorly characterized. Current evidence cannot distinguish causal relationships from shared risk factors, creating a critical knowledge gap for developing effective prevention strategies.
To address this gap, we will employ advanced genetic methodologies within the UK Biobank cohort. Our specific aims are to: (1) develop and validate polygenic risk scores for both diseases using a two-stage approach; (2) quantify shared genetic architecture via cross-trait PRS associations and pleiotropic locus identification; and (3) determine bidirectional causality using Mendelian randomization to establish evidence for causal pathways.
Genetic instruments enable causal inference in complex diseases. Polygenic risk scores integrate genetic susceptibility across variants to quantify disease risk, while Mendelian randomization utilizes these variants as natural experiments to establish causality and overcome confounding. Together, these methods provide powerful tools to elucidate disease etiology beyond conventional observational approaches. To solve this, our study will use genetic data as a powerful tool to uncover the true relationship. We will develop polygenic risk scores, which measure a person’s inherited risk for each disease. Using a method called Mendelian randomization, we will then treat these genetic scores as natural experiments to test if higher genetic risk for stroke actually causes a higher risk of Alzheimer’s, or if the reverse is true. The findings will provide crucial evidence on whether preventing or treating cerebrovascular disease can also reduce the risk of developing Alzheimer’s dementia, guiding future clinical strategies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-uk-biobank-longitudinal-study-of-clonal-haematopoiesis-and-risk-of-alzheimers-disease

A UK Biobank longitudinal study of clonal haematopoiesis and risk of Alzheimer’s Disease

Last updated:
ID:
81793
Start date:
20 April 2022
Project status:
Current
Principal investigator:
Dr Burcu Cevik
Lead institution:
Ankara University, Turkiye

Considering the causes of Alzheimer’s disease, hereditary factors come to the fore for Early Onset Alzheimer’s Disease. However, the causes of Late-Onset Alzheimer’s Disease, which constitutes the majority of the patient population, have not been clarified yet. Age is the most prominent risk factor for Alzheimer’s Disease, but the effects of ageing on the disease are not fully understood. With ageing, mutations which are not acquired congenitally occur in blood cells. These mutations have been found to be less than 1% in people with age >40, about 10% in people with age >70, and about 15-20% in people with age >90 in investigations involving people without haematological malignancy. This condition has been termed clonal haematopoiesis of indeterminate potential. Individuals with these mutations are at risk of developing haematological malignancies such as Myelodysplastic Syndrome, Acute Myeloid Leukaemia, and Lymphoma. In addition, these mutations increase the risk of coronary heart disease, myocardial infarction and mortality. It has been discovered that these mutations, the prevalence of which increases with ageing, lead to some gene activations and inflammatory effects. We intend to use Whole Exome Sequencing data from the UK Biobank to investigate whether clonal haematopoiesis of indeterminate potential is a risk factor for Alzheimer’s disease in this study. The findings of this project will shed light on the aetiology and pathogenesis of Alzheimer’s disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-unified-approach-to-the-holistic-detection-of-prevalent-diseases

A Unified Approach to the Holistic Detection of Prevalent Diseases

Last updated:
ID:
921607
Start date:
17 September 2025
Project status:
Current
Principal investigator:
Professor Dimitris Bertsimas
Lead institution:
Massachusetts Institute of Technology, United States of America

Cancer, cardiovascular disease, diabetes (types 1 and 2), neurological disorders, and kidney disease present vast health and economic challenges. It is thus important to diagnose diseases early and tailor treatments precisely to the diverse needs of patients. In addition, no major disease exists in isolation, motivating a holistic understanding of the biology of each person.

We seek to use UK Biobank data to (i) identify molecular and physiological biomarkers predictive of disease predisposition, onset, and progression, and (ii) uncover cross-disease biosignatures that reveal shared biological connections.

Under Dr. Dimitris Bertsimas, our group has a proven track record in health AI. We demonstrated the strengths of holistic machine learning (ML) approaches, (i) developing a ML framework that integrates multimodal data (e.g. labs, notes, images) to reliably predict patient outcomes (Integrated Multimodal Artificial Intelligence Framework for Healthcare Applications, Soenksen et al., 2022), (ii) extending this to multitask across multiple clinical endpoints (M3H: Multimodal Multitask Machine Learning for Healthcare, Bertsimas & Ma, 2023), and (iii) writing an ML textbook on improving healthcare delivery (The Analytics Edge in Healthcare, Bertsimas, Orfanoudaki, & Wiberg, 2023).

We believe we can incorporate cellular-level data with outpatient visit data to create a more comprehensive and holistic biological picture of each patient. We aim to identify biosignatures for early, accurate, and minimally invasive disease detection, by integrating multiomics data and clinical phenotypes to identify clinical-molecular associations that can better model disease trajectories.

Through this unified ML approach, we aim to build interpretable data-driven models that inform early detection and personalized treatment strategies. Our expertise and experience uniquely position us to uncover both shared and distinct etiologies across these critical disease categories.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-validation-study-using-uk-biobank-for-discovery-driven-analysis-of-temporal-disease-progression-patterns-using-gcat-a-catalan-linked-electronic-health-registry-cohort

A validation study using UK Biobank for discovery-driven analysis of temporal disease progression patterns using GCAT, a Catalan linked electronic health registry cohort.

Last updated:
ID:
97879
Start date:
31 May 2023
Project status:
Current
Principal investigator:
Dr Rafael de Cid
Lead institution:
Germans Trias i Pujol Research Institute (IGTP), Spain

Human disease, is a complex trait that occurs once in a life, based on a biological basis, however its expression is variable, on intensity, onset and duration, because its personal genetic basis (gene-centred and genome-wide) and their environment. In this point one disease itself or mediated by secondary effects of it treatment, modifies the presentation of other conditions, that also based on their genetics (shared or not) and their environment, modify the occurrence of further diseases or their evolution. This is the rule for most of common diseases, chronic conditions that occurs once and that are treated chronically.
Our research plan is based on the time and order in which diseases are diagnosed in the population: First, we will apply different technical approaches to identify trajectories based on single diagnostics or aggregation analysis using cluster comorbidities, based on age-gender strata. Second, using genetic profiles (genome wide), we will analyse shared genetics among members of trajectories, o lead disease among clusters, to identify polygenic profiles that could identify temporary progression. PRS will be used as Instrumental variables. Adjusted variables will be used to account for impact of ethnic and educational level on disease diagnostics. Third, UKBB, will be used as a validation steep, using the same approach used in the Catalan cohort, to validate the results. The increased power of the UKBB database, will allow the exploration of additional observed comorbidities or temporary patterns that due to lack of power in the discovery cohort could not be explored in the first steep.
The identification of the genetic profile, as biomarkers or as etiological determinants, will help us gain a better understanding of the diseases and the health of the population, moving to an informed practice of the health-care management. This will impact the health-care system, with a preventive and more personalized management of diseases using agnostic genetic information. In addition, promising disease relationships would be derived for further investigated using more in depth approached to identify pleiotropic loci, as a step for a better drug repositioning strategy.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/a-vision-of-healthy-urban-design-for-ncd-prevention

A vision of healthy urban design for NCD prevention

Last updated:
ID:
65425
Start date:
1 February 2021
Project status:
Current
Principal investigator:
Dr Ruth Hunter
Lead institution:
Queen's University Belfast, Great Britain

Non-communicable diseases (NCD), such as cancer, heart disease, type II diabetes mellitus, chronic respiratory conditions like asthma, and poor mental health, are some of the most common causes of death in the UK. The design of our cities play an important role in preventing these chronic diseases and have a significant consequent impact on the quality of life and life expectancy of their citizens. However, in a rapidly evolving urban age, urban designers, urban planners and public health practitioners still know surprisingly little about how best to design our cities in order to prevent NCD and their known risk factors.
Our aim is to generate evidence and tools to support the urban planning and health sectors to better understand how to design our cities to prevent NCD. Objectives include:
1. Use new methods in computer vision and artificial intelligence to explore the relation between urban design and NCD in cities across the UK.
2. Investigate how different designs within cities impact on health inequalities including NCD.
3. Combine data from different sources to investigate the mechanisms by which the design of our cities causes NCD.
4. Learn lessons about how different ways of designing our cities prevent NCD and their known risk factors.
5. Develop a toolkit for action for local citizens, urban designers and planners, public health practitioners and policy makers, to help inform future policies and lead to powerful, actionable changes in the city.
6. To build a legacy of transdisciplinary research capacity in public health science, urban design and computer science, with clear pathways to impact.
We will build the evidence base on the relationship between urban design and NCD. This will be comprised of two levels of analysis: the city level and individual level. This acknowledges that people’s health is affected by their immediate environment and by the way the entire city is organised. This will help us understand which features of the built environment are associated with city designs, NCD and their known risk factors, such as not being physically active, having poor diet, smoking, consuming alcohol and being exposed to air pollution. We will then test changes to the design of our cities to analyse how they will help prevent NCD and reduce known risk factors. The findings will help inform future policies and practices leading to actionable changes in the design of our cities to make them healthier places for people to live.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/abdominal-obesity-and-diabetic-kidney-disease-a-mendelian-randomization-study

Abdominal obesity and Diabetic Kidney Disease: A Mendelian Randomization Study

Last updated:
ID:
66536
Start date:
26 October 2020
Project status:
Current
Principal investigator:
Dr Jinbo Hu
Lead institution:
The First Affiliated Hospital of Chongqing Medical University, China

Aim: Diabetic kidney disease (DKD) develops in approximately 40% of patients who are diabetic and is the leading cause of end stage renal disease worldwide. Our previous study indicated that abdominal obesity is an important risk factor of DKD, consistent with several observational studies. However, the causal correlation between the abdominal obesity and DKD remains unclear.

Scientific rational: To identify the causal correlation between the abdominal obesity and DKD, we intend to use the approach of Mendelian randomization, which has traditionally been used to determine whether a candidate risk factor is causally related to a clinical outcome using the genotype as an instrument. In a Mendelian randomization study, genetic variants is used as to divide a population into genotypic subgroups, in an analogous way as how participants are divided into arms in a randomized controlled trial. By dividing participants into subgroups with different genetically exposures to the risk factor, we can evaluate the causal effect of the risk factor on disease risk. A recent large-scale Genome wide association study (GWAS) identified 48 SNPs associated with abdominal obesity, and combining these 48 SNPs into a weighted polygenic risk score, we can determine whether a genetic predisposition to abdominal obesity is associated with DKD.

Project duration: The duration of this project will be three years, from 2020 to 2023.

Public health impact: We expected to identify the causal association between abdominal obesity and the development of DKD. If further research validates our findings, two recommendations on the risk assessment and intervention of DKD might be considered by clinical practice: (1) Clinical screening of abdominal obesity by simple anthropometric measures to identify diabetic patients who might be more susceptible to diabetic nephropathy; (2) Interventions aiming to reduce abdominal adipose tissue may become an important adjunct to glycemic and blood pressure control for reducing the risk of DKD.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/accumulated-environmental-factors-on-sleep-mental-health-and-cognitive-outcomes

Accumulated Environmental Factors on Sleep, Mental Health, and Cognitive Outcomes

Last updated:
ID:
755949
Start date:
17 June 2025
Project status:
Current
Principal investigator:
Dr Junxin Li
Lead institution:
Johns Hopkins University, United States of America

Research on environmental influences on cognition/ADRD has primarily focused on single exposures, suggesting air pollution, noise , limited greenspace , sunlight, and artificial nightlight exposure, are individually associated with heightened risks of ADRD, mental health problems, and sleep disturbances. In real world settings, however, people are often exposed to multiple adverse environmental factors simultaneously. These overlapping exposures may amplify health risks beyond the effects of individual factors. Understanding the aggregated impact of these exposures is essential for identifying high-risk combinations and evaluating the combined effects of multiple exposures on sleep and cognitive health in aging. This study will introduce an aggregated environmental exposure model to identify high-risk environmental combinations for sleep, mental health and AD/ADRD.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/achieving-personalized-medicine-in-cardiometabolic-diseases-based-on-heterogeneity-in-treatment-response-defined-by-novel-proteomic-biomarkers

Achieving personalized medicine in cardiometabolic diseases based on heterogeneity in treatment response defined by novel proteomic biomarkers

Last updated:
ID:
316056
Start date:
14 April 2025
Project status:
Current
Principal investigator:
Dr Itsuki Osawa
Lead institution:
Columbia University, United States of America

It is critically important to prescribe the right medications at the right time to get the best treatment results for patients. Our previous research has found ways to improve treatment strategies by understanding how different patients respond to the same treatments. This was done using information like patient demographics, medical history, and standard lab results. However, because cardiometabolic diseases (like hypertension, type 2 diabetes, and heart disease) have complex underlying causes, this clinical information alone may not be enough to fully understand a patient’s condition. To better personalize treatments, we plan to use advanced protein analysis (proteomics) to: 1) identify how different patients respond to treatments for cardiometabolic diseases, 2) understand the molecular reasons behind these different responses, and 3) determine the best use of each drug based on new proteomic biomarkers found in blood samples. By combining traditional clinical data with proteomic data, we hope to more accurately assess patient conditions and tailor treatments to improve outcomes.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/acquired-risk-factors-and-incidence-of-metabolic-cardiovascular-disease-according-to-age-and-genetic-susceptibility

Acquired Risk Factors and Incidence of Metabolic Cardiovascular Disease According to age and Genetic Susceptibility

Last updated:
ID:
151350
Start date:
24 October 2024
Project status:
Current
Principal investigator:
Dr Jianmin Yang
Lead institution:
Qilu Hospital of Shandong University, China

The social environment, dietary habits, lifestyle, clinical comorbidities, and body metabolism play a key role in the global burden of metabolic cardiovascular disease. Therefore, identification and effective management of modifiable factors are essential for the prevention and control of CVD. Given the differences between populations, the importance of various risk factors varies across populations. Therefore, by utilising data from the UK Biobank, we plan to calculate group attributable risks for metabolic CVD risk factors in different age and genetic susceptibility groups. Subsequently, we will rank these risk factors according to group attributable risk to identify high-risk, modifiable factors. In addition, we will investigate how metabolic cardiovascular risk factors vary with individual age and genetic susceptibility. This study will build on existing evidence and support national and international guidelines for early intervention in the management of metabolic cardiovascular disease. Understanding the interactions between genetics and metabolic cardiovascular risk factors will contribute to more precise management of metabolic cardiovascular disease in the future.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/actigraphic-sleep-patterns-of-restless-legs-syndrome

Actigraphic Sleep Patterns of Restless Legs Syndrome

Last updated:
ID:
759578
Start date:
5 August 2025
Project status:
Current
Principal investigator:
Dr John Weyl Winkelman
Lead institution:
Massachusetts General Hospital, United States of America

Background:
Restless Legs Syndrome (RLS) is a neurological disorder characterized by discomfort in the legs while resting and an uncontrollable urge to move the legs to relieve the discomfort. RLS more typically occurs in older adults. RLS commonly follows a circadian rhythm, worsening in the evening and at night, delaying sleep onset. Thus, while older adults without RLS typically fall asleep and wake up earlier than younger adults, those with RLS often fall asleep later and have their best sleep in the early morning. This distinct pattern of sleep has been anecdotally noted by Dr. John Winkelman, MD, PhD.

We aim to compare sleep patterns of RLS and non-RLS patients, as measured by hour-by-hour actigraphic data. If a unique pattern of sleep is identified, distinguishing RLS patients from non-RLS patients, it can be used as a tool to aid in the diagnosis of Restless Legs Syndrome.

Research Question:
How do the sleep patterns of patients with Restless Legs Syndrome (RLS) differ from those without the condition, based on actigraphic measurements?

Objectives:
– Compare actigraphic data of sleep patterns in RLS patients and non-RLS controls.
– Investigate if RLS patients exhibit a distinct sleep pattern, particularly in the early hours of the night and morning.

Scientific Rationale:
Although the pattern of sleep disturbance in RLS has been noted anecdotally, there has been little systematic investigation into these circadian trends, especially using objective, quantifiable measures like actigraphy. This project aims to fill this gap by using actigraphy to compare the sleep patterns of RLS patients versus non-RLS controls, offering insight into the possible use of sleep disturbances as a diagnostic feature for RLS.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/activity-data-as-an-input-to-epsrc-funded-project-the-wearable-clinic-connecting-health-self-and-care

Activity data as an input to EPSRC funded project ?The Wearable Clinic: Connecting Health, Self and Care?

Last updated:
ID:
33693
Start date:
6 August 2018
Project status:
Closed
Principal investigator:
Professor Alexander Casson
Lead institution:
University of Manchester, Great Britain

We aim to enable new forms of collaborative care for long-term conditions by integrating data from wearable sensors (accelerometery) with the prediction of health risks from electronic health records. This will let us create algorithms for adaptive, personalized care planning that takes account of individual predicted risks and real-time sensing. We will investigate serious mental illness (schizophrenia), chronic kidney (renal) disease, and controls. We will quantify episodes of activity data as different measures/risk factors, use this as an input to our predictive modeling, and contributes toward an understanding of whether activity monitoring can be used an indicator of remission/relapse. We are developing software tools which will help patients with long-term conditions, together with their carers and doctors, to better manage their health in daily life, respond more quickly to changes, and prevent fall back episodes. By identifying associations between unsupervised behavioral phenotype (mobility, rhythmic/routines, sedentary behavior, fitness/frailty issues, weight gain/loss) and disease progression stages, we will build new disease risk prediction models. We have a strong focus on translation, with a dedicated 3 year work package on health and economic benefit analysis, so we can advance diagnosis, prevention/early detection, and risk stratification for chronic kidney disease and schizophrenia. The analysis stage will identify indicators (i.e. patterns/phenotype/risk factors) in the accelerometer data that assign patients to different disease states.(?Unsupervised? machine learning methods will be used to identify patterns in the data itself, without any intervention from the user, or a human interpreter.) These accelerometer based indicators will be used as one input, together with data on hospitalizations, self-reported measures, blood samples, and similar, for developing risk prediction models of how likely each person is to relapse, or to be in remission, or to be in another identifiable state which might alter their care planning.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/activity-tracking-and-whole-body-mri-for-the-prediction-of-individual-wellness-comparative-analysis-with-chinese-cohorts

Activity Tracking and Whole-Body MRI For The Prediction of Individual Wellness -Comparative Analysis with Chinese Cohorts

Last updated:
ID:
78730
Start date:
1 February 2022
Project status:
Current
Principal investigator:
Dr Varut Vardhanabhuti
Lead institution:
University of Hong Kong, Hong Kong

Chronic diseases such as diabetes, cardiovascular diseases, and cancers develop through multiple culmination of processes including lifestyle, dietary, environmental and genetic factors. The development of diseases occur on a continuum with the final stages resulting in detectable disease, and patients having symptoms. There are several stages prior to this, that we can detect early changes in the body, in which if intervention can be targeted, can be more effective at disease prevention and cure. This study will examine the early changes based on body MRI quantitative measurements and physical activity measurements. These changes can be subtle and will require comparison with population reference ranges which can now be calculated based on the data collected by the UK Biobank.

Our project aims to derive population normative range using the UK biobank data and the local Chinese cohorts with data collected in the same manner and to combine the various quantitative parameters for assessing individual wellness.

The idea is we may be able to create biomarkers for the cardiovascular, cancer-related and all-cause mortality prediction. The impact to the broader population will be that there will be a more accurate method of screening of subclinical disease detection based on these non-invasive data points opening a window for serial monitoring during the intervention to improve health and individual wellness. Accurate and reproducible biomarkers are desirable not only to improve how we can measure health but also in assisting large scale clinical trials. We anticipate the findings to significantly contribute to the advancement of preventative medicine. The project duration is expected to be 36 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/addressing-metabolic-and-neuroinflammatory-factors-for-disease-progression-in-pd

Addressing metabolic and neuroinflammatory factors for disease progression in PD

Last updated:
ID:
435848
Start date:
12 June 2025
Project status:
Current
Principal investigator:
Dr Stanislav Groppa
Lead institution:
Universitatea de Stat de Medicina si Farmacie "N. Testemitanu", Moldova (Republic of)

Parkinson’s disease (PD), is one of the most debilitating neurodegenerative disorders characterised by motor symptoms including bradykinesia, tremor, and rigidity. While a complex interaction of genetic and molecular mechanisms has been postulated to play an important role for the vulnerability of brain networks to neurodegeneration, little is known about the metabolic and neuroinflammatory mechanisms that substantially modify disease courses. Recent work using single-cell RNA transcriptomes from cortical tissue of Parkinson’s patients has identified a strong association between disease progression subtypes and subsets of microglia, oligodendrocyte progenitors and astrocytes. Dysregulated mitochondrial electron transport, abnormal oxidative phosphorylation pathways and an inflammatory microglia response might be mainly involved. In this project we aim defining disease progression phenotypes (defined as age at disease onset, motor impairment at distinct disease durations and motor and cognitive worsening over disease course) to proteomic and metabolomic characteristics. We will then relate these to distinct charactersitics of neurons and glia subsets as derived from own sc-transcriptomes. Genetic and MRI variables will be considered as confound factors and further included in the multivariable analyses. We aim describing novel direct mechanistic links between neuroinflammation and neurodegeneration through mutual cellular subsets and gene proteomic and metabolomic expressions in PD for different disease subtypes can be targeted in future personalised therapeutic interventions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/adherence-to-cancer-prevention-recommendations-and-risk-of-in-situ-breast-cancer

Adherence to cancer prevention recommendations and risk of in situ breast cancer

Last updated:
ID:
55149
Start date:
23 March 2020
Project status:
Closed
Principal investigator:
Professor Sabine Rohrmann
Lead institution:
University of Zurich, Switzerland

Few studies have investigated potential risk factors for in situ breast cancer . Although the association between in situ breast cancer risk and few individual risk factors has been investigated, analyses on diet and lifestyle in relation to in situ breast cancer from large studies are missing.

The World Cancer Research Fund/American Institute for Cancer Research has recently published nine cancer prevention recommendations. Adherence to these recommendations has been associated with reduced cancer incidence and mortality, including reduced invasive breast cancer risk. However, to our knowledge, no study has investigated the association between adherence to the World Cancer Research Fund/American Institute for Cancer Research cancer prevention recommendations and in situ breast cancer risk.

In the analysis anticipated, each of the potentially modifiable World Cancer Research Fund/American Institute for Cancer Research cancer prevention recommendation will be scored, as in previous publications. The sum of the single recommendations will then constitute the score used to assess the risk of in situ breast cancer development. Since in situ breast cancer is mainly detected through mammographic screening, we will utilize the mammographic information in the UK Biobank (mammographic screening attendance, time since last mammogram) as covariates and stratifying variables in our analyses.

We aim to investigate whether adherence to the potentially modifiable World Cancer Research Fund/American Institute for Cancer Research cancer prevention recommendations is associated with in situ breast cancer development overall and by socioeconomic characteristics in the UK.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/adherence-to-dietary-patterns-genetic-predisposition-and-incidence-of-coronary-artery-disease

Adherence to dietary patterns, genetic predisposition and incidence of coronary artery disease

Last updated:
ID:
44033
Start date:
29 May 2019
Project status:
Closed
Principal investigator:
Dr Romina di Giuseppe
Lead institution:
University Hospital Schleswig-Holstein, Germany

Coronary artery disease (CAD) is a complex traits with heritable, environmental, and lifestyle factors contributing to disease risk and disease course. Both, genetic as well as dietary factors are known to be associated with risk for new-onset (incident) CAD. However, data on the interplay of dietary patterns (a more comprehensive analytical approach to diet than single food analyses) and genetic variants (cumulatively expressed as genetic risk score) on the risk of incident CAD are lacking. We are applying for data from the UK Biobank to address this issue. Specifically, we are aiming to:
1. Derive a priori (Alternate Mediterranean Diet, Alternate Healthy Eating Index-2010, and the Dietary Approach to Stop Hypertension) and a posteriori (using PFA, PCA, RRR, and LCA) dietary patterns based on data from the UK Biobank;
2. Assess associations between these derived dietary patterns and incident CAD – as defined in (46) – within the UK biobank.
3. Assess whether the association of dietary patterns with incident CAD differs by genetic risk (based on a weighted genetic risk score).
4. Investigate influences of scoring alternatives on CAD risk.
5. Investigate changes of dietary patterns over time and their determinants and assess how these changes relate to CAD status alone and according CAD-GRS categories.
The present project will increase our understanding of the complex interplay between genetic factors and nutrition with respect to incident CAD. It might inform future personalized heart-nutrition strategies aimed at preventing CAD.
Full cohort required. Data request: FFQ and FFQ repeated measures, web-based 24-hour-recall questionnaires, clinical, anthropometric and CVD biomarkers data, family history of CAD, genetic data, CAD endpoints.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/adiposity-measurements-types-of-body-fat-weight-change-and-related-genetic-markers-in-relation-to-cancer-risk

Adiposity measurements, types of body fat, weight change, and related genetic markers in relation to cancer risk

Last updated:
ID:
24487
Start date:
1 March 2017
Project status:
Closed
Principal investigator:
Dr Wei Zheng
Lead institution:
Vanderbilt University Medical Center, United States of America

Virtually no epidemiologic studies have evaluated the association of objectively measured central obesity with cancer risk. The UK Biobank cohort provides an exceptional opportunity to evaluate this association. We propose: 1) to evaluate the associations of objectively measured body fats, weight gain, and BMI with total and site-specific cancer risk; 2) to determine associations of BMI and central obesity with breast and colorectal risks via Mendelian randomization analyses; and 3) to assess the association between GWAS-identified BMI-associated genetic variants and weight change over the lifecourse. The associations of both adiposity and adult weight gain with cancer risk are of increasing public health importance because the prevalence of overweight and obese adults has increased markedly in recent decades. The results of our proposed study will increase knowledge of the influence of obesity on cancer risk. Additionally, our Mendelian randomization study, along with the evaluation of BMI-associated variants with weight gain, will provide insight into the complex relationship of genetically-determined body weight and cancer risk. We will use statistical methods to evaluate the association of cancer risk with body fat, fat around the mid-section, and weight change from childhood to adulthood. We will evaluate these relationships using information collected on body composition during the participants? baseline visits, and by using information from genetic markers that are known to be related to body mass index. We plan to conduct a cohort analysis for the first aim, including all cohort members who provided the data needed for the analysis. For the second aim, a nested case-control study will include all cases of breast (2,320) and colorectal (1,256) cancer and 4 matched controls per case whose genotyping data are available (approximately 17,880 total). The third aim will utilize all participants with sufficient genotyping data, a baseline measure of BMI, and at least one of the following measurements: a value for comparative body size at age 10 or a repeat assessment value for BMI.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/admission-uk-multimorbidity-research-collaborative-on-multiple-long-term-conditions-from-burden-and-inequalities-to-underlying-mechanisms

ADMISSION UK Multimorbidity Research Collaborative on Multiple Long-Term Conditions: from burden and inequalities to underlying mechanisms

Last updated:
ID:
73744
Start date:
7 March 2022
Project status:
Current
Principal investigator:
Professor Heather Jane Cordell
Lead institution:
Newcastle University, Great Britain

Living with multiple long-term health conditions is common and important for both patients and for the NHS (National Health Service). The way we deliver health services in the NHS is not ideally suited to care for people with multiple long-term conditions. To prevent people from acquiring multiple long-term conditions and to provide the best care to people with multiple long-term conditions we urgently need to better understand how to prevent and treat different groups of long-term health conditions that tend to occur together, known as clusters of long-term conditions.

As part of our 4 year collaborative programme of research, funded by the Medical Research Council and National Institute for Health Research we plan to use data from UK Biobank to address this need by:
1) establishing how the area where a person lives and other socio-demographic factors are linked with different clusters of long-term conditions. We will also look at whether lifestyle factors such as diet and exercise that may have a role in explaining any links we find
2) gaining new understanding of what processes lead to the development of different clusters of long-term conditions, using the genetic data available in UK Biobank.

This work will lead to the development of robust evidence that will provide the basis for novel public health, policy and biopsychosocial interventions to prevent, treat and mitigate the consequences of different clusters of long-term conditions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/adult-cancer-risk-among-those-who-were-breast-fed-when-they-were-babies

Adult cancer risk among those who were breast-fed when they were babies

Last updated:
ID:
25417
Start date:
1 April 2017
Project status:
Closed
Principal investigator:
Dr TienYu Owen Yang
Lead institution:
University of Oxford, Great Britain

It is hypothesised that carcinogenic viruses could be transmitted through breast milk and cause breast cancer. There might be other differences between babies fed with breast milk and with bottle milk that contribute to cancer in adulthood, such growth hormones or gut microorganisms. Whether there is difference in adult cancer risk between babies who are breast-fed and were bottle-fed remains under-investigated. We propose to compare risk of adult cancer risk and related diseases between those who were breast-fed and those who were not breast-fed when they were babies among UK Biobank participants. One purpose of the UK Biobank is to ?support a diverse range of research intended to improve the prevention, diagnosis and treatment of illness and the promotion of health throughout society.? The proposed research aims to understand whether breast feeding is associated with cancer risk of specific sites and risk of related diseases. The results may provide hints to preventable causes of cancer. At the recruitment of the UK Biobank, participants reported a wide range of information, including whether they were breast-fed when they were babies. They also gave consent so that their ongoing history of hospital admissions and cancer diagnosis after recruitment could be obtained for research. Among participants who did not have cancer at the recruitment, we will compare risk of developing newly diagnosed cancer and related diseases between participants who reported they were breast-fed and not breast-fed when they were babies in the UK Biobank. The full cohort is requested for this study.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/advanced-imaging-pipelines-to-further-our-understanding-of-how-genotype-the-brain-and-personal-history-interact

Advanced imaging pipelines to further our understanding of how genotype, the brain, and personal history interact.

Last updated:
ID:
63956
Start date:
16 June 2021
Project status:
Current
Principal investigator:
Dr Lee Lancashire
Lead institution:
Cohen Veterans Bioscience, United States of America

Genetics, socioeconomic status, education, neurobiology and medical history all interact in complex ways to confer sensitivity or resilience to central nervous system insult. For example, even mild TBI, in individuals with high CNS frailty, may lead to increased risk for stroke, neurodegenerative disease, anxiety or depression later in life. The traumatic brain injury (TBI) research group at the University of Virginia, in collaboration with Cohen Veterans Bioscience, proposes the use and analysis of the neuroimaging, genetic, cognitive testing, medical history, and other demographical data available within the UK Biobank repository to study the linkage between neurological and cardiovascular health to better understand the diverse outcomes (i.e. the sub-populations) represented in military veterans and other aging populations. These data and resulting analyses will inform potential precision medicine approaches, guide future research avenues, and further refine the development of open-source tools to conduct such research.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/advanced-machine-learning-techniques-for-early-detection-and-prediction-of-prediabetes-in-healthcare

Advanced Machine Learning Techniques for Early Detection and Prediction of Prediabetes in Healthcare

Last updated:
ID:
332898
Start date:
4 December 2024
Project status:
Current
Principal investigator:
Mr Mahmoud BA Almadhoun
Lead institution:
Universiti Teknikal Malaysia Melaka, Malaysia

The goal of this research project is to develop and refine machine learning models to better identify individuals at risk of developing diabetes. Specifically, the project aims to:
1. Detect individuals with prediabetes.
2. Stratify these individuals into low, intermediate, and high-risk categories for progressing to diabetes.
Scientific Rationale:
Prediabetes is a condition where blood sugar levels are higher than normal but not yet high enough to be classified as diabetes. Early detection of prediabetes is crucial because it allows for timely interventions that can prevent or delay the onset of diabetes. Diabetes is a serious condition that can lead to severe health complications such as heart disease, kidney failure, and blindness.Traditional methods of diagnosing and assessing the risk of diabetes have limitations, such as relying on a few specific biomarkers and not accounting for the complex interactions between different risk factors. Machine learning, a type of artificial intelligence, can analyze large amounts of data and identify patterns that might not be apparent to human researchers. By leveraging electronic health records and comprehensive datasets, we can develop more accurate and personalized models for predicting who is at risk of progressing from prediabetes to diabetes.
Project Duration:
The project is expected to last three years, including the phases of data collection, model development, validation, and implementation.
Public Health Impact:
This research has the potential to significantly improve public health by:
1. Enhancing Early Detection:** More accurately identifying individuals with prediabetes, allowing for earlier and more effective intervention strategies.
2. Personalizing Care:** Stratifying individuals based on their risk levels helps tailor prevention and treatment plans, making them more effective.
3. Reducing Healthcare Costs:** By preventing the progression to diabetes, the project can help reduce the long-term healthcare costs associated with treating diabetes and its complications.
4. **Improving Health Outcomes:** Ultimately, the research aims to improve the quality of life for individuals at risk of diabetes by preventing the onset of the disease and its associated health issues.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/advanced-omics-non-omic-determinants-and-phenotyping-for-cardiovascular-disease-and-comorbidities

Advanced Omics, Non-Omic Determinants, and Phenotyping for Cardiovascular Disease and Comorbidities

Last updated:
ID:
339480
Start date:
5 November 2024
Project status:
Current
Principal investigator:
Dr Chuangshi Wang
Lead institution:
Fuwai Hospital Chinese Academy of Medical Sciences, China

Cardiovascular diseases, such as ischemic heart disease, hypertension, stroke, heart failure, peripheral artery disease, etc., are among the leading causes of mortality among adults. Identifying risk factors for these diseases is crucial for preventing or delaying complications and premature death.
Aims:
1. To comprehensively assess proteomic, metabolomic, genomic, and other omic factors, as well as imaging, environmental, lifestyle, psychosocial factors, etc., within the UK Biobank and their causal associations with cardiovascular diseases and comorbidities.
2. To evaluate the associations between trajectory changes in omic and non-omic risk factors, with the risk and mortality of cardiovascular diseases and multi-morbidity.
3. To develop risk prediction tools (e.g., models, scores, algorithms, etc.) that incorporate omic and non-omic factors for cardiovascular diseases and multi-morbidity.
4. To utilize advanced statistical methods (e.g., machine learning) to improve the accuracy and efficiency of identification and risk prediction of cardiovascular diseases and their comorbidities.
Scientific Rationale: Although numerous factors affecting the risk of cardiovascular diseases have been identified, many more remain undiscovered. Non-omic factors, including dietary habits, exercise behavior, sleep patterns, nutritional status, etc., are well known to influence these diseases. Additionally, the interactions between proteomic, metabolomic, genomic, and non-omic factors must be considered. Moreover, there is a lack of sufficient and robust evidence on estimating the risk and mortality of cardiovascular diseases by incorporating the trajectory changes of these factors in a population-based prospective cohort. The integration of advanced statistical methods and phenotyping techniques can further enhance the identification and prediction of cardiovascular diseases by uncovering complex patterns and relationships in the data. A better understanding of the factors affecting the likelihood of developing cardiovascular diseases is essential for creating novel and more effective approaches to disease prevention, diagnosis, and treatment. This project is expected to last for 36 months.
In addition, several other diseases (including metabolic, respiratory, digestive, neurological, renal diseases, and cancers) are known to have significant links and interactions with cardiovascular health. By comprehensively analyzing data on these related diseases, we can better inform the development of improved prevention strategies, targeted interventions, and personalized treatments for individuals at risk of multi-comorbidity.
Public Health Impact: Our study will enhance the understanding of the etiology of cardiovascular diseases and provide scientific evidence for early intervention and improved disease management. This will ultimately contribute to better public health outcomes by informing strategies to reduce the burden of cardiovascular diseases and their comorbidities.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/advanced-predictive-models-for-health-risk-assessment-and-estimating-health-intervention-effectiveness-insights-from-the-uk-biobank

Advanced Predictive Models for Health Risk Assessment and Estimating Health Intervention Effectiveness: Insights from the UK Biobank

Last updated:
ID:
281730
Start date:
22 October 2024
Project status:
Current
Principal investigator:
Dr Abbas Salami
Lead institution:
University of Essex, Great Britain

Our project combines computer science and public health expertise to explore and enhance health risk prediction models using data from the UK Biobank. We aim to understand better the complex relationships between lifestyle choices, environmental factors, and other variables and how they impact health outcomes such as mortality, diseases, hospital visits, and cardiovascular events. Traditional health risk assessments often look at isolated factors without fully accounting for how these factors interact. Our approach seeks to fill this gap by analysing the interconnections among various health risk factors, thus improving our ability to estimate their combined effects on individual and population health.
Concurrently, we are investigating the effectiveness of a gamified digital health intervention through a separate, ongoing Randomised Controlled Trial (RCT). This trial examines how gamification, implemented via the YuLife mobile health app, can motivate users to adopt healthier behaviours. The insights gained from the UK Biobank will complement the findings from the trial, enabling us to assess how lifestyle modifications influenced by gamification impact significant health outcomes. Our research aims to identify the risk factors most responsive to gamified digital interventions and predict the long-term impact of these interventions on health. This is particularly relevant in today’s world, where lifestyle-related chronic diseases are becoming more common. By combining the comprehensive biomedical data of the UK Biobank with the practical interventions tested in the YuLife app, we hope to establish a solid scientific basis for developing engaging, effective, and personalised public health strategies. The ultimate goal of our two-year project is to develop actionable, evidence-based recommendations that can be used by health practitioners, policymakers, and individuals to reduce health risks and enhance quality of life. These recommendations will focus on identifying and implementing effective and accessible digital health intervention strategies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/advanced-risk-prediction-and-heritability-estimation-methods

Advanced Risk Prediction and Heritability Estimation Methods

Last updated:
ID:
56885
Start date:
30 April 2020
Project status:
Current
Principal investigator:
Professor Malka Gorfine
Lead institution:
Tel Aviv University, Israel

In this proposal we focus on estimating the risk of developing the disease (e.g. breast cancer) over time given risk factors. Survival analysis is a branch of statistics for analysing the time until a pre-specified event happen, such as death or a disease. Hence, it provides a useful tool for such prediction analysis while taking into account competing risks (i.e., a person who died before having the disease cannot have the disease anymore), and left truncation (i.e., only individuals who survive at least by age 40 are included in the UK Biobank dataset). Heritability summarise the proportion of the variance of the trait under study (e.g. disease status) that is due to genetic factors. Predictive performance of a model strongly depends on the extent of heritability of the trait. For any given sample size, more accurate prediction is possible for more heritable traits, such as Crohn disease and type 1 diabetes, than for less heritable traits such as prostate cancer. Improving heritability estimation for various traits could provide insights on several missing heritability questions and lead to practical solutions for improving risk-prediction models.
Finally, we will study complex and high dimensional traits. Current methods, which follow a two-step approaches: first, extract the relevant features from the high-dimensional traits and then perform genome-wide association analysis on these features. In contrast to these methods, we will apply a joint modelling approach where we extract the most heritable features by correlating them with the genotype data, which will enable us to discover new heritable traits, and may lead to changes in definition of relevant traits from high-throughput digital trait data.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/advanced-statistical-and-machine-learning-approaches-for-identifying-gene-by-environment-interactions-across-ancestral-populations

Advanced Statistical and Machine Learning Approaches for Identifying Gene-by-Environment Interactions Across Ancestral Populations

Last updated:
ID:
66192
Start date:
17 January 2022
Project status:
Current
Principal investigator:
Dr Curtis Malik Boykin
Lead institution:
Brown University, United States of America

Rationale:
The sequencing of the human genome offered a lot of promise for understanding the genetic basis behind many traits that are consequential for health and wellness. By understanding the genome, we might be able to understand why certain individuals suffer from specific illnesses, which could offer insight into how to manage and treat certain diseases. While decades of research have revealed a very strong genetic basis for many traits, questions remain about how certain aspects of our environment or experience may play a role in how we study genomes. More advanced statistical techniques may allow us to better measure how the environment may influence disease risk.

Aim:
We aim to study how many under-appreciated features of the environment-including those associated with social stratification (e.g., socioeconomic status)—can influence how we study and interpret the relationship between genetics and observable traits. We will develop and apply new statistical methods across a number of traits and environmental factors, learning how certain environmental factors may interact with and affect the genetic architecture of behavioral and psychological traits. The UK Biobank contains hundreds of thousands of individuals from diverse demographic backgrounds, which makes it an ideal set to examine these questions. It will allow us to examine many traits of interest and examine different environmental factors. Gene-by-environment interactions have been well studied in other complex traits (e.g., height). We plan to evaluate the power and robustness of our developed methods on these phenotypes, and then focus on behavioral and psychological traits, which can be influenced by experiences, and contribute to mental health conditions.

Duration:
Because the foundation for the statistical methods that we will apply for this work are published, we anticipate a brief study time. We anticipate 12 months from the start of the study, to study completion, where analyses will be complete, with the team starting the process of sharing results with the scientific community via publication shortly thereafter.

Public health impact:
While the science of genomics has already provided insight into how genes can influence disease risk, there remain many other factors that can influence disease risk. For example, the lived experience and exposure to certain environments can lead to certain mental health ailments. Our study hopes to propose better methods for separating the effects of genes from those of the environment. Further, we hope to identify some specific factors that might make some individuals more susceptible to mental illness.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/advancing-actigraphy-based-daily-activity-and-sleep-analysis-with-machine-learning-to-understand-baseline-biometrics-in-healthy-individuals-and-those-experiencing-various-medical-conditions

Advancing actigraphy-based daily activity and sleep analysis with machine learning to understand baseline biometrics in healthy individuals and those experiencing various medical conditions

Last updated:
ID:
45113
Start date:
18 December 2018
Project status:
Closed
Principal investigator:
Dr Jian Wang
Lead institution:
Eli Lilly and Company Ltd (USA), United States of America

While physical activity and good sleep is beneficial for human health both physically and mentally, most evidence of such claim is based on self-assessed measures of activity. In the UK Biobank study, data from activity monitors are collected 24/7 for 7 days from over 100K participants producing over 15 trillion movement readings. Coupled with extensive demographic and health information, this dataset will give us the first glimpse of baseline biometrics from healthy individuals as well as individuals experiencing various medical conditions.

Conventional algorithms to detect sleep from actigraphy data require static sleep diaries or actigraphy marker button to label the boundaries of time in bed. These manual practices add to the uncertainty and participant burden, which we aim to reduce by automating detection of sleep boundaries during both night sleep and day time napping from the data directly. Here, we aim to explore probabilistic models for sleep detection to improve upon the conventional algorithms that only distinguish binary sleep versus waking time. Finally, we aim to translate these innovations into open source software feasible to be re-used by non-domain experts.

Collaborating with Dr. Vincent van Hees, the Netherlands eScience Center in Amsterdam, the Netherland, we will begin a two year study to develop the algorithm and apply it in UK Bioback dataset to derive sleep and activity metrics in medical conditions to serve as benchmark for Lilly clinical studies planning to incorporate activity monitor as quality of life assessment tool.

Leveraging public datasets collected in the last few years by large observational studies such as the Whitehall II Study (Sabia et al., 2014), UK Biobank (Doherty et al., 2017), and the Newcastle 85+ study (Anderson et al., 2014), the project would advance the activity monitor data open source software development and accelerate public and private efforts to study impact of sleep health and physical activity on healthy living, aging, disease, as well as transition from wellness to illness, and possibly illness back to wellness with the aid of lifestyle adjustment or therapeutics


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/advancing-cardiovascular-risk-identification-with-structured-clinical-documentation-and-biosignal-derived-phenotypes-synthesis-acribis

Advancing Cardiovascular Risk Identification with Structured Clinical Documentation and Biosignal Derived Phenotypes Synthesis (ACRIBiS)

Last updated:
ID:
155031
Start date:
14 February 2024
Project status:
Current
Principal investigator:
Professor Christoph Dieterich
Lead institution:
Heidelberg University Hospital, Germany

Personalized risk assessment is recommended in cardiovascular medicine guidelines. Scores are often used to individually adapt prevention, diagnosis and treatment. However, the implementation of this risk assessment in everyday clinical practice in Germany is inadequate due to major obstacles: the lack of relevant structured information; inadequately standardized and incomplete storage in electronic health records; Lack of interfaces and data linkage to enable rapid assessment and ultimately visualization. The same applies to high-resolution biosignal analysis, which represents an important untapped resource for risk assessment. We are researching novel concepts to combine score and biosignal-based risk analyzes and define their predictive performance in real clinical settings.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/advancing-cmml-research-through-the-integration-of-genomic-and-phenotypic-data

Advancing CMML Research through the Integration of Genomic and Phenotypic Data.

Last updated:
ID:
233227
Start date:
27 November 2024
Project status:
Current
Principal investigator:
Mr Umesh Padia
Lead institution:
Massachusetts Institute of Technology, United States of America

Chronic myelomonocytic leukemia (CMML) is a rare and aggressive blood cancer that affects the production of certain white blood cells called monocytes. Patients with CMML often have a poor prognosis, with limited treatment options and a high risk of progression to more severe forms of leukemia. Despite recent advances in our understanding of the molecular changes associated with CMML, the underlying genetic causes of the disease remain largely unknown. This lack of knowledge has hindered the development of effective prevention, diagnosis, and treatment strategies for CMML.

Our research project aims to address this challenge by leveraging the resources of the UK Biobank, which is a large-scale biomedical database containing genetic and health information from 500,000 individuals. By comparing the data of individuals with CMML to those of healthy individuals in the UK Biobank, we hope to identify specific variations that may contribute to the development and progression of CMML.

To achieve this goal, we will employ state-of-the-art computational methods to analyze the vast amounts of genetic and health data available in the UK Biobank. This will involve using machine learning algorithms to identify patterns and associations between genetic variations and CMML risk, as well as developing predictive models that can assess an individual’s likelihood of developing the disease based on their genetic profile, biomarkers, proteins, and other health factors.

The expected duration of our research project is three years. During this time, we will work closely with clinical collaborators and patient advocacy groups to ensure that our findings are clinically relevant and can be translated into meaningful benefits for patients with CMML.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/advancing-complex-traits-understanding-through-multidimensional-data-integration-and-analysis

Advancing Complex Traits Understanding through Multidimensional Data Integration and Analysis

Last updated:
ID:
172174
Start date:
28 October 2024
Project status:
Current
Principal investigator:
Jinlong Shi
Lead institution:
The General Hospital of the People's Liberation Army, China

Complex traits and diseases typically arise from the interplay of various elements, such as genetic, environmental, and lifestyle factors. Effectively addressing these diseases often demands a comprehensive approach that takes into account genetic predisposition, lifestyle modifications, and, when necessary, medical interventions. The rapid advancement of today’s technology is instrumental in studying complex traits using high-dimensional datasets, offering innovative frameworks to unravel the intricate connections between biomolecules, biological phenomena, and the environment. These approaches have been applied to numerous clinical studies, advancing disease diagnosis and enhancing treatment options for patients.
The UK Biobank stands as a treasure trove of data, encompassing genetic and lifestyle information about a substantial population. By integrating and analyzing extensive data from the UK biobank and our own collections, we aim to gain deep insight into the genetic basis of complex traits and establish related prevention and control measures.
We seek to 1) investigate the underlying mechanisms of complex traits and the intricate interactions among multiple factors. 2) explore the relationship between complex traits and other disorders. 3) intergrate imaging phenotypes with multi-omic biological data to develop sophisticated deep learning systems and models. These will aid in the accurate identification of clinical subtypes or drug resistance, prediction of effective treatment strategy, and identification of predictive biomarkers.
Project duration: Estimated duration of this project is expected to be 3 years.
Output: Our intended output over the next 2-3 years is to identify several biomarkers or reveal mechanisms that could lead to 1-2 publications in prestigious journals such as Nature Genetics, Nature Medicine, and Nature Communications.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/advancing-early-detection-and-mechanistic-understanding-of-neurological-critical-diseases-through-large-scale-cohort-data

Advancing Early Detection and Mechanistic Understanding of Neurological Critical Diseases through Large-Scale Cohort Data

Last updated:
ID:
1023951
Start date:
28 September 2025
Project status:
Current
Principal investigator:
Miss Zhihan Zhang
Lead institution:
Tianjin University, China

Research questions
This research aims to advance the early detection and mechanistic understanding of neurological critical diseases, as well as related cardio-cerebrovascular and systemic conditions, by addressing the following aspects:
1. Identifying key clinical, biological, electrophysiological, genetic, and lifestyle factors associated with the onset, progression, and outcomes of neurological critical diseases
2. Developing deep learning models to predict disease risk, early manifestations, and critical events using large-scale, multi-dimensional cohort data.
3.Utilizing large language models (LLMs) to generate automated, personalized diagnostic and prognostic reports, making predictive insights accessible and clinically actionable.
Research objectives
1. Develop predictive models using deep learning and diverse cohort data (electrophysiological signals, genetics, biomarkers, imaging, clinical records) to identify early indicators of neurological critical and cardiovascular diseases.
2. Improve risk stratification by integrating lifestyle, medical history, and multi-modal biomarkers with primary data sources.
3. Automate clinical reporting with LLMs to generate personalized diagnostic and prognostic reports, supporting efficient and informed care.
scientific rationale for the research
Neurological, cardiovascular, cerebrovascular, and sleep-related diseases are major causes of global morbidity and mortality, yet early detection remains limited. The UK Biobank offers large-scale, high-quality data from over 500,000 participants, including electrophysiological signals, genetics, imaging, biomarkers, and clinical records. This project will apply deep learning to identify early indicators, improve risk stratification, and generate mechanistic insights. Large language models will translate predictive outputs into automated reports, supporting precision medicine and reducing the public health burden of these diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/advancing-genomic-research-enhancing-gwas-and-twas-through-comprehensive-data-integration-in-the-uk-biobank

Advancing Genomic Research: Enhancing GWAS and TWAS through Comprehensive Data Integration in the UK Biobank

Last updated:
ID:
129007
Start date:
3 January 2024
Project status:
Current
Principal investigator:
Professor Pedro Beltrao
Lead institution:
ETH Zurich, Switzerland

We will investigate how variation in the genome influences diseases in the UK Biobank dataset. To understand how diseases work and what genes and proteins affect a human disease or trait, researchers search for relations between gene mutations and diseases. While currently, researchers look for associations of genomic areas or genes with traits in GWAS and TWAS methods, the results are not always accurate. One reason for the inaccuracies might be that a predicted change in mRNA abundance does not necessarily cause a change in protein abundance. So, taking this into account, to improve and extend these approaches, we plan to integrate additional data into the studies, mainly proteomics data. That will elevate the current results by providing another set of associations: between proteins and traits. The method will allow us to study the UK Biobank data from a different perspective and compare the protein-level associations with the ones from GWAS studies to prioritize and complement meaningful relationships between genetics, proteomics, and disease. We will carry out the research project on the entire set of UK Biobank samples. With this, we hope to provide important insights into disease and trait mechanics


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/advancing-genomics-for-diverse-and-admixed-populations

Advancing Genomics for Diverse and Admixed Populations

Last updated:
ID:
95179
Start date:
21 March 2024
Project status:
Current
Principal investigator:
Dr Elizabeth Grace Atkinson
Lead institution:
Baylor College of Medicine, United States of America

In recent years, genomic studies have been able to identify genetic variants associated with important complex traits which can inform precision medicine. However, these studies have mainly included people of European ancestry, and individuals of other ancestry are still greatly underrepresented in genetic research. Diversifying genomic studies is critical to ensure that the benefits of these findings are shared beyond European populations.

We aim to build and test tools to better study the genetics of complex traits in diverse populations. This addresses two main barriers that currently prevent broader inclusion in genomics: the lack of analytical methods that are tailored to diverse ancestries, and the lack of analyses conducted that include diverse populations. Our work will improve the clinical utility of large-scale data-collection efforts and begin to address the concerning health disparities that exist across populations.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/advancing-knowledge-of-diseases-through-genotype-to-phenotype-translational-research-for-medical-student-research-training

Advancing Knowledge of Diseases through Genotype to Phenotype Translational Research For Medical Student Research Training

Last updated:
ID:
92550
Start date:
29 May 2025
Project status:
Current
Principal investigator:
Professor Mohanakrishnan Sathyamoorthy
Lead institution:
Texas Christian University, United States of America

Our project aims to advance scientific knowledge by studying the association between patient specific characteristics in disease states and underlying associations with novel genetic mutations. The disease states we plan to study include aneurysms, arrhythmias, cardiomyopathies, and clotting disorders. The key aim of these projects will be to verify novel discoveries across a much larger sample, as contained in the UK Biobank. In the medical and scientific community, this type of research is known as phenotype to genotype correlation and has been well established as a method to advance knowledge of disease states.

The public benefit of our research may eventually lead to enhanced diagnosis, and more appropriate treatments of the conditions which we will study.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/advancing-knowledge-on-physical-activity-and-health-through-investigating-interrelationships-with-lifestyle-behaviours-fitness-and-subclinical-outcomes

Advancing knowledge on physical activity and health through investigating interrelationships with lifestyle behaviours, fitness, and subclinical outcomes

Last updated:
ID:
29717
Start date:
6 April 2018
Project status:
Current
Principal investigator:
Dr Ulf Ekelund
Lead institution:
Norwegian School of Sport Sciences, Norway

The overall aim is to examine the independent and combined associations between obesity, MVPA, sedentary time and CRF with all-cause, cardio-vascular and cancer mortality.
1. To examine the independent and combined associations of MVPA, obesity, sedentary time and CRF associated with mortality?
2. To examine whether physical activity modify the association between obesity (adiposity) and mortality?
3. To examine whether sedentary time mediate the association between obesity and mortality? Obesity, moderate and vigorous intensity physical activity (MVPA), sedentary time and cardio-respiratory fitness (CRF) are all associated with a number of various health outcomes and mortality. However, the combined associations between these lifestyle variables and mortality is unknown. Further, it is unknown whether objectively measured PA may modify the association between adiposity (BMI and waist circumference) on mortality and whether objectively measured sedentary time may mediate this association. We will quantify how obesity, physical activity sedentary time and fitness are associated with mortality and the relative importance of physical inactivity, obesity, high sedentary time and low fitness on mortality. This will enable important information for intervention strategies and policy makers. Full cohort n=500,000 for most research questions, n~100,000 for the analyses involving cardio-respiratory fitness and objectively measured physical activity.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/advancing-personalised-medicine-for-psychiatric-diseases-through-integrative-gpcr-pharmacogenomics

Advancing personalised medicine for psychiatric diseases through integrative GPCR pharmacogenomics

Last updated:
ID:
55955
Start date:
24 June 2020
Project status:
Current
Principal investigator:
Dr Alexander Sebastian Hauser
Lead institution:
University of Copenhagen, Denmark

Understanding how drug response to a given therapeutic treatment differs in patients is one of the most important and long-standing challenges in personalised medicine. For example, 30 – 50% of patients with major depression do not respond to their first antidepressant drug prescription, and adherence remains often very poor. While a great deal of effort has been placed on linking variation in drug transport and metabolism with treatment responses, very little is known about how genetic variability in receptor drug targets affects treatment response. Elucidating the spectrum and impact of how a genetic receptor variant influences ligand binding and/or signalling and, ultimately, therapeutic effect is vital and would serve as an important step for understanding variability in drug response.

The G protein-coupled receptor (GPCR) super-family spanning ~800 members (~4% of the human genome) allows environmental and physiological messages – communicated by distinct signalling molecules – to be relayed into adequate cellular responses. Apart from involvement in almost all physiological processes, GPCRs also mediate the effect of ~34% of drugs (Hauser et al., 2017 Nat Rev Drug Discov). In this project we focus on drugs used to treat schizophrenia, affective disorders and attention deficit hyperactivity disorder (ADHD), which prevalently target GPCRs directly or indirectly. Despite the significance of GPCRs and genetic variations for CNS drug therapies, personalised medicine is yet beyond reach, because we are missing integrated large-scale data showing which genetic missense variants affect the intended therapeutic effects and how.

Here, we will employ a unique ‘pharmacogenomics’ research strategy, which integrates receptor genetic variants with 3D structures, pharmacological experiments, patient registries and literature data. We first map and select genetic variants in GPCR drug targets affecting CNS drug response through computational models. We then determine how psychiatric drug responses are affected by genetic receptor target variations in cell-based assays. Those experiments are labor-intensive, as we need to capture all possible intracellular responses. We hence envisage that the project would approximately take 3 years. We expect to correlate similar molecular receptor phenotypes induced by genetic variation with clusters of prescription changes and reported adverse reactions. To test these hypotheses, we will integrate information on hospitalisation records and prescribed medicines within the UKBioBank cohort.

A successful project would provide evidence for genetic variants linked to therapies and would hence allow us to:

i) better understand treatment adverse reactions in psychiatric disorders
ii) personalise medicine prescriptions based on GPCR genotypes, and
iii) prioritise drugs for pharmacovigilance investigations


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/advancing-understanding-of-multi-morbidity-in-metabolic-disease-through-innovation-in-statistical-machine-learning

Advancing understanding of multi-morbidity in metabolic disease through innovation in statistical machine learning

Last updated:
ID:
83855
Start date:
14 September 2022
Project status:
Current
Principal investigator:
Dr David Jenkins
Lead institution:
University of Manchester, Great Britain

Many people have more than one long-term disease (known as multimorbidity). For instance, there are many people who have both heart disease and diabetes. There exist computer tools that help doctors to predict the risk that a patient will develop diseases in the future (for instance, heart disease). However, these tools always focus on single diseases. This is not helpful when we try to prevent and treat multimorbidity. In this project we will develop tools that predict the risks that a patient will develop multiple diseases (and what those diseases are).

Working out how best to treat patients with multimorbidity is not straightforward. Typically, each disease comes with its own course of treatment, but it is not clear how to combine these. Therefore, we will also develop tools that predict what would happen if a patient were given a particular treatment. Such a treatment could involve changing their lifestyle (e.g. stop smoking), taking a particular drug, or a combination of these things.

People with multimorbidity often end up taking many drugs (known as polypharmacy). This can be a problem because some drugs do not work when they are taken with other drugs. We will therefore also extend the computer tools such that they can predict what would happen if a patient, who is living with multimorbidity and is taking multiple drugs, would change their lifestyle or change the drugs that they are taking.

We will focus on patients with diabetes, heart disease, and related diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/advancing-understanding-of-psychiatric-disorders-via-genetic-analyses

Advancing understanding of psychiatric disorders via genetic analyses

Last updated:
ID:
102638
Start date:
15 May 2023
Project status:
Current
Principal investigator:
Professor Anders Børglum
Lead institution:
Aarhus University, Denmark

Psychiatric disorders are a significant public health problem, accounting for a significant portion of disabilities worldwide. Studies have shown that many of these disorders have a strong genetic component, with heritability estimates for disorders like schizophrenia and autism being as high as 80%. Despite this, the genetics of these conditions are still not fully understood, with the majority of genetic risk factors yet to be identified.

This project aims to better understand how genetics affects psychiatric and neurodevelopmental disorders, with a particular focus on autism, ADHD, bipolar disorder, major depressive disorder, schizophrenia, and substance use disorder. To achieve this, we will apply state-of-the-art analytical approaches to data from the UK Biobank and Danish population-based case-cohort sample for genome-wide association studies (GWAS), whole-exome sequencing (WES) and whole-genome sequencing (WGS) studies. The goal is to identify novel genetic risks and advance our knowledge of the genetic architecture of these disorders, with a focus on the role of common and rare genetic variants, genetic differences in sex, and genetic differences across subgroups.

The long-term goal of this project is to improve the diagnosis and treatment of psychiatric disorders by utilizing the knowledge gained from these studies. To achieve this, we aim to construct a prediction model that incorporates various types of data such as genetic data, electronic health records (EHR), imaging data, and environmental data. By doing so, we hope to improve the accuracy of prediction and facilitate the early detection of psychiatric disorders, particularly when they exhibit similar signs and symptoms.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/advancing-understanding-prediction-prevention-and-treatment-of-coronary-heart-disease-through-application-of-genetics-to-uk-biobank

Advancing understanding, prediction, prevention and treatment of coronary heart disease through application of genetics to UK Biobank

Last updated:
ID:
9922
Start date:
1 February 2015
Project status:
Closed
Principal investigator:
Dr Christopher Paul Nelson
Lead institution:
University of Leicester, Great Britain

Coronary heart disease (CHD) and heart attack is caused by a combination of inherited and lifestyle/environmental factors. 17,000 new cases of CHD are expected to develop in the UK Biobank participants by 2017. The aims of our research are use the genetic data being generated in UK Biobank to identify genes that affect risk of CHD, investigate whether specific genes and certain lifestyle factors such as smoking interact to increase risk, determine whether adding genetic information can improve prediction of CHD, and identify causal mechanisms for CHD that can be targeted to develop new treatments. UK Biobank was established to improve understanding of the causes of common diseases and particularly the interaction between genes and environment and life-style factors. CHD is the commonest cause of death and disability world-wide. It is the archetypal disease caused by an interaction between inherited and environmental/lifestyle factors. Our research will improve understanding of the genetic causes of CHD, how these interact with environmental/lifestyle factors and how the findings can be applied to improving prediction, prevention and treatment of CHD. We will divide the UK Biobank participants into those with CHD (cases) and those without (controls) and compare their genetic information generated using the UK Biobank array to identify genetic variants that are associated with development of CHD. We will use lifestyle/environmental information collected on participants to see if there is an interaction between such factors (e.g smoking) and genes in increasing CHD risk. We will investigate whether adding genetic information to current methods can improve prediction of development of CHD. We will use genetic approaches to define the best targets to develop drugs against to tackle CAD We plan to use the full cohort for our studies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/advancing-understanding-prediction-prevention-and-treatment-of-the-comorbidities-through-genetics-applications-at-uk-biobank

Advancing understanding, prediction, prevention and treatment of the comorbidities through genetics applications at UK Biobank.

Last updated:
ID:
246093
Start date:
11 November 2024
Project status:
Current
Principal investigator:
Dr Min Yi
Lead institution:
Guangzhou Medical University, China

Comorbidity is commonly defined as at least two conditions coexisting in the same individual, which has posed a great challenge to the healthcare system worldwide and created substantial effect on individuals, their families, and society. The mechanisms of comorbidities are closely intertwined, with multidirectional relationships, shared risk factors, and common therapeutic targets. These results demonstrate the need for a comprehensive assessment of comorbidities to establish comprehensive disease models to promote the development of precision medicine.
However, the shared genetic architecture and biological mechanisms underlying comorbidities development are still unclear. Most previous studies have focused on observational studies, which were affected by confounding factors and cannot reflect the direct causal relationship between exposure and outcomes. In addition, most of these researches mainly focus on three elements: clinical characteristics, auxiliary examination results, and demographic characteristics, ignoring the impact of genetic factors on the disease. Therefore, it is necessary to further explore shared genetic architecture and deeper mechanisms of the comorbidities, which may contribute to the understanding, prediction, prevention, and treatment of the diseases.
This proposal aims to explore how comorbidities affects each other using Mendelian randomization design which is less susceptible to confounding than observational studies. We also aim to find some potential drug targets through the investigation of genetic architecture and their biological mechanisms and to apply them to the prediction, prevention, treatment, and management of diseases.
The project will last for 36 months. This proposal aims to investigate the causal association among comorbidities with each other using Mendelian randomization instrumental variable analysis. The findings will provide additional insights concerning the bi-directional association among comorbidities, which may improve our understanding of the pathophysiological mechanisms and help identify novel biological pathways and therapeutic targets for improving the prediction, prevention, and treatment of the diseases. By improving the diagnosis and treatment of co-morbidities, people can be encouraged to seek timely medical help, and the financial burden on the healthcare system and society can be released.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/adverse-childhood-experiences-and-analgesic-outcomes-in-adults-living-with-chronic-pain-and-multimorbidity

Adverse childhood experiences and analgesic outcomes in adults living with chronic pain and multimorbidity.

Last updated:
ID:
107405
Start date:
10 November 2023
Project status:
Current
Principal investigator:
Dr Dhaneesha Senaratne
Lead institution:
University of Dundee, Great Britain

Why we are doing this research: Adverse childhood experiences (ACEs) are stressful events that happen to people before the age of 18. They include things like abuse, neglect and parental separation. Previous research has shown that experiencing more ACEs as a child leads to poorer health as an adult. This might be because specific conditions (like chronic pain) are more severe. Or it might be because people who have experienced ACEs develop multiple long-term health conditions that are hard to deal with at the same time. We want to test the theory that people who have had ACEs may respond differently to pain medications (such as morphine), compared with those who have not had an ACE.

Our approach: People with lived experience of ACEs, chronic pain and multiple long-term health conditions helped us to decide on our research questions. This helps us ensure our work can explore topics that are meaningful to the relevant people.

Some example questions:
Compared with people who have not experienced ACEs, are people who have experienced ACEs more likely to…
1) … be prescribed pain medications?
2) … be prescribed higher doses of pain medications?
3) … experience harms relating to pain medications (such as addiction, overdose or death)?
4) … find pain medications unhelpful?
5) … have side-effects from pain medications?
We will see if the answers to these questions are different for people with multiple long-term health conditions. We will use the UK Biobank database to answer these questions. We think that the large size of this database (over half a million people) will make our results relevant to patients and clinicians.

Project duration: 36 months.

Public health impact: Knowing how ACEs affect our responses to pain medications is important. It may help some people understand how their childhood affects their current health. For many people, knowing “why” can help them come to terms with their condition. This research will highlight the importance of trauma-informed care, and may guide patients and healthcare professionals when discussing treatment plans. It may be that for some people pain medications don’t help, and that other options (such as psychological therapy) might be better.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/adverse-health-consequences-of-a-sitting-occupation

Adverse health consequences of a sitting occupation

Last updated:
ID:
30585
Start date:
8 December 2021
Project status:
Current
Principal investigator:
Professor Chris Dibben
Lead institution:
University of Edinburgh, Great Britain

Prolonged sitting can have serious health consequences and a high level of fitness does not protect against the negative effects of sitting for too long. Sedentary job poses therefore a risk to all individuals regardless of their physical activity outside work. We aim to identify occupations at high risk of prolonged sitting and advance our understanding of the health risks associated with sitting for extended periods of time. We will evaluate the impact of lengthy sitting on the risk of developing cardiovascular disease and/or type 2 diabetes. A score evaluating risk of prolonged sitting for a given occupation will be derived for future research. Self-reported and objective accelerometer-measured sedentary behaviour data will be used to examine health risks associated with prolonged sitting. Sitting in the workplace is of primary interest but different sedentary behaviours and their interactions with physical activity will be taken into account, with adjustment for socio-demographic, genetic, and behavioural factors. We expect this project to take 24 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/adverse-outcomes-of-cardiovascular-drugs

Adverse outcomes of cardiovascular drugs

Last updated:
ID:
105044
Start date:
7 July 2023
Project status:
Current
Principal investigator:
Professor Jinzhu Jia
Lead institution:
Peking University, China

Cardiovascular drugs, such as aspirin and statins, are effective in preventing and treating cardiovascular disease (e.g. heart attacks and stroke). However, their potential effect on other chronic diseases (e.g. cancer, osteoporosis, respiratory diseases, dementia, and Alzheimer’s disease) remains unclear. The aim of this project is to investigate the effect of cardiovascular drugs on complex chronic diseases. We will use data from the full UK Biobank cohort. And innovative statistical methods, such as matching method and negative control method, will be used to increase the credibility of results. This study is expected to last for 36 months. The findings will provide evidence-based guidance for healthy cardiovascular drug use.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/adverse-pregnancy-outcomes-and-cardiovascular-disease-risk

Adverse Pregnancy Outcomes and Cardiovascular Disease Risk

Last updated:
ID:
99869
Start date:
9 June 2023
Project status:
Current
Principal investigator:
Dr Svati Shah
Lead institution:
Duke University, United States of America

Aims:
We aim to use the data available from the UK Biobank to understand how genetic, metabolic, and clinical factors affect the intersection between adverse pregnancy outcomes and cardiovascular risk in women.
Background:
The genome is made up of 3 billion base pairs – A, C, T & G – and variation in the genetic code has been identified to be associated with risk for many diseases. Certain cardiometabolic traits and medication target loci have been associated with risk for hypertensive disorders of pregnancy. Expanding off of this, it will be valuable to look across many other APO (phenotypes) to determine other important associations that may exist between cardiometabolic traits and medication target loci with all APOs, not just hypertensive disorders of pregnancy.
Duration:
The anticipated project should take approximately three years after obtaining access to the data.
Public health impact:
Despite advances in genetic and other molecular techniques, determination of which patients are at most risk of for a broad array of APOs has yet to be explored. This project may potentially identify novel associations of diseases associated with the genetic, metabolomic, and clinical areas we plan to focus on. Ultimately, we hope this work will lead to more personalized approaches to pregnancy and postpartum care and improved maternal morbidity outcomes. Understanding the interaction between APOs and CV risk will help us to identify women at risk, better estimate their long-term CV risk, and improve our efforts to prevent and manage heart disease aggressively.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/aetiology-and-prediction-of-cardiometabolic-diseases-their-comorbidities-and-complications

Aetiology and prediction of cardiometabolic diseases, their comorbidities and complications

Last updated:
ID:
55469
Start date:
28 February 2020
Project status:
Current
Principal investigator:
Dr Michael Inouye
Lead institution:
Baker Heart and Diabetes Institute, Australia

Cardiometabolic diseases are the leading cause of death and morbidity worldwide. In number of deaths, they are followed by their complications, such as kidney disease, heart attack and stroke as well as neurological conditions, such as Alzheimer’s disease. Together, they are responsible for the most deaths and the highest costs in the world in terms of treatments and hospitalisation. These conditions are caused by both genetics (inherited DNA) and environment (diet, activity, sleep).

In our research we aim to investigate (i) critical genes, regulatory regions and their interactions that influence risk of the disease; (ii) the complex interplay, between the genes and environment, that underpins both health, and risk of developing diseases; (iii) understand the causal mechanisms, biomarkers and progression of disease in order to develop new and improved treatments; and (iv) determine genetic risk of metabolic diseases.

We will use the genetic information and wide variety of disease and other phenotypes available in UK Biobank, such as biomarkers and imaging data.

Despite advances in prediction as well as changes in lifestyle and medication these diseases are still on the rise globally. Key challenges in the prevention and management are to: (i) understand better the complex molecular pathways and mechanisms leading to their development, progression, and complications; (ii) determine the causal relation of biomarkers and other putative risk factors, and (ii) find new therapeutic approaches to reduce deaths due to those disorders.

Our proposal will (i) improve understanding of the genetic mechanisms of cardiometabolic and neurological diseases and their comorbidities; (ii) provide a better understanding of how genetics interact with environmental factors in development and progression of these conditions; (iii) and potentially lead to improving their prediction, prevention and treatment.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/african-ancestry-genomics-of-drug-response-and-disease-risk

African Ancestry genomics of drug response and disease risk.

Last updated:
ID:
67216
Start date:
8 March 2021
Project status:
Current
Principal investigator:
Dr Minoli Perera
Lead institution:
Northwestern University, United States of America

SUMMARY:
DNA Difference between people may be able to predict if they will develop diseases or respond to medications. However, there is a lack of studies to find these important predictors in people of African Ancestry. We have collected a large cohort of patients with heart problems in which we have collect DNA, how well they respond to medications for their heart problems, and what heart conditions they currently suffer from. We are using this data to find if the predictors in African Ancestry patients are different from what we already know in European patients. This could lead to new diagnostic tests that can be used by doctor to diagnoses and treat patient of African descent.

AIMS:
1) Better understand how genomics and clinical factors influence drug response in African Ancestry patients
2) Better understand how genomics and clinical factors predict risk of disease in African Ancestry patients

DURATION:
We are initially applying for a project duration of 36 months, with a option to extending if studies are ongoing.

PUBLIC HEATH IMPACT:
The studies proposed will generate knowledge in the field of pharmacogenomics (the study of how genetics affect response to drugs) and disease genomics (determining if genetics increases the risk on developing a disease), with the ultimate goal of improving outcomes for African ancestry patients.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/after-the-plague-genetic-history-of-medieval-cambridge-population-in-relation-to-present-day-population-of-britain

After the plague: genetic history of medieval Cambridge population in relation to present-day population of Britain

Last updated:
ID:
54698
Start date:
17 February 2020
Project status:
Closed
Principal investigator:
Professor Toomas Kivisild
Lead institution:
Katholieke Universiteit Leuven, Belgium

This project studies the historical effects of disease epidemics and social stratification on the population of Cambridge through time. Using evidence from ancient and modern DNA as well as methods from archaeology, history, osteoarchaeology, isotopic and osteopathology it asks how healthy and how different from each other were people from different social classes in the past and how they compare to the people from different parts of Britain today, using the UK Biobank data as a reference. One of the main foci of the project is on a recently excavated large sample of urban poor people from the Hospital of St. John (AD 1200-1500), complemented by comparative samples from other medieval social contexts and other historical periods. The results will be analysed both statistically and biographically. A proximate goal is to build a nuanced picture of health, lifestyle and activity in medieval England, one grounded in direct examination of human bodies themselves. The overall goal, however, is to understand the biosocial effects of the Black Death of 1348-1350, an epidemic of bubonic plague which decimated Europe. By comparing samples from before and after the epidemic for a wide range of social and biological indicators, including phenotype related evidence from the UK Biobank, this research will aim to reveal how the plague changed human well-being, activity, mobility, health and the genetic constitution of Europe.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/age-and-gender-adjusted-low-serum-apolipoprotein-a1-high-c-reactive-protein-as-predisposing-biomarkers-of-sars-cov-2-infection-a-large-retrospective-study-using-uk-biobank

Age and gender adjusted low serum apolipoprotein A1, high C-reactive protein as predisposing biomarkers of SARS-CoV-2 infection. A large retrospective study using UK biobank.

Last updated:
ID:
79974
Start date:
18 March 2022
Project status:
Current
Principal investigator:
Professor Thierry Poynard
Lead institution:
Assistance Publique - Hôpitaux de Paris, France

The aim of this project is to validate a blood based risk stratification tool, combining existing and routinely available markers (apolipoprotein-a1, C-reactive protein) with age and gender that could be an additional diagnosis and prognosis tool for screening subjects at high-risk of SARS-Cov-2 infection (SI).

The 2 biomarkers have a published role in the SARS-Cov-2 infection, with normal values depending on age and gender. A score combining the values of the 2 assays, corrected by age and gender, will be correlated to the risk of SI.

The project will last 3 years.

The public health impact could be major, helping the clinicians stratifying the patients according to this novel risk score.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/age-and-risk-adjusted-trajectories-of-retinal-neural-loss-in-uk-biobank-using-deep-learning-rnfl-from-fundus-photos-benchmarked-to-oct-with-genetic-metabolomic-and-epigenetic-modifiers

Age and Risk Adjusted Trajectories of Retinal Neural Loss in UK Biobank Using Deep-Learning RNFL from Fundus Photos, Benchmarked to OCT, with Genetic, Metabolomic and Epigenetic Modifiers

Last updated:
ID:
1021268
Start date:
4 November 2025
Project status:
Current
Principal investigator:
Dr Felipe Andrade Medeiros
Lead institution:
University of Miami, United States of America

This project investigates ocular and systemic determinants of retinal health and disease risk.
Loss of retinal neurons is a hallmark of ocular ageing and a key contributor to vision loss from diseases such as glaucoma. It may also mirror brain health, offering a non-invasive window into neurodegeneration. However, there is limited large-scale evidence quantifying how retinal nerve fiber layer (RNFL) metrics change across adulthood, and which lifestyle, systemic, and genetic factors accelerate or protect against thinning.
Our goals are to: (1) quantify age-related trajectories of RNFL measures across adulthood, assessing possible non-linearities; (2) identify ocular and systemic risk factors (e.g., blood pressure, metabolic markers, sleep, activity, air pollution exposure, epigenetic clocks) associated with faster RNFL thinning; (3) evaluate genetic contributions using polygenic risk scores and exome/WGS summary metrics; and (4) relate RNFL measures to diagnosed eye and systemic diseases using linked records.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/age-associated-proteomic-signatures-and-their-role-in-age-related-co-morbidities

Age-associated proteomic signatures and their role in age-related co-morbidities

Last updated:
ID:
489309
Start date:
13 February 2025
Project status:
Current
Principal investigator:
Dr Ozlem Bulut
Lead institution:
Radboud University Medical Centre, Netherlands

Using the OLINK platform’s inflammation and cardiovascular panels, we have constructed a proteomic age score and calculated proteomic age acceleration (how much proteomic age differs from the chronological age) using data from a Dutch cohort (n=302) established at Radboudumc. However, this and other cohorts that we have access to lack follow-up information. With the UK Biobank data, we aim to understand if the proteomic age acceleration is able to predict cardiovascular events and/or mortality and how it compares to classical risk factors in terms of prediction. We would like to calculate the proteomic age score of the UK Biobank participants for which OLINK measurements are available and to test the predictive power using Cox proportional hazard regression.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/age-estimation-and-cardiac-disease-prediction-from-ppg-signals-using-interpretable-machine-learning-models

Age estimation and cardiac disease prediction from PPG signals using interpretable machine learning models.

Last updated:
ID:
549696
Start date:
19 February 2025
Project status:
Current
Principal investigator:
Professor Cynthia Rudin
Lead institution:
Duke University, United States of America

Our study aims to develop a new, more accurate way of predicting heart health by using interpretable machine learning to assess biological aging. Unlike chronological age, which only reflects the years a person has lived, biological age can provide insights into the actual health of their cardiovascular system. For this, we will analyze Photoplethysmography (PPG) signals-light-based measurements often used to monitor blood flow-to estimate how a person’s heart and blood vessels are aging.
The goals of this research are threefold: (1) Create an interpretable machine learning model that can predict a patient’s cardiovascular age accurately, (2) Identify and explain the specific biological factors influencing these age predictions, and (3) Investigate the relationship between chronological age and biological age to better understand how they align or differ in the context of heart health.
The scientific rationale behind this study is to bridge the gap between chronological and biological age measurements. Many people with the same chronological age can have vastly different cardiovascular health, which could mean that some individuals are at risk of heart disease sooner than expected. By focusing on biological age, we hope to provide a more personalized assessment of cardiovascular health, leading to better preventative care.
We plan to disseminate out finding through manuscript publication, and the release of our model and code in a publicly accessible repository.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/age-prediction-from-neuroimaging-and-genetic-profiles

Age Prediction from NeuroImaging and Genetic Profiles

Last updated:
ID:
55274
Start date:
3 March 2020
Project status:
Current
Principal investigator:
Ethan MacDonald
Lead institution:
University of Calgary, Canada

Understanding of the aging process is critical to understanding functional declines and diseases associated with aging such as cardiovascular disease and cancer. In this analysis study, our aim is to improve the understanding of how various features change during the aging process and to construct a mathematical model which can be used to determine how old a person is biologically. This would provide a tool which would allow for investigation of the influence of various factors on the aging process and to flag outliners which have a large deviation of the estimated age and chronological age.
While age estimation has been done and there are many new applications published in the literature this year. However, model calibration with the UK BioBank database is relatively limited to date, and due to its size we expect it to perform well and allow for more advanced model types. Being able to estimate the age of a person allows us to get a sense of their overall health.
In Canada, there are supercomputing resources available for University employed researchers, with greater than 58,000 cores for performing experiments just like this. Such a system is required to run image analysis jobs of this scale and to answer the important questions proposed. A project of this nature is generally tedious to implement as it can take a long to transfer data, arrange and structure the data for such analysis. Furthermore, a project of this nature is often iterative as a processing pipeline of this nature takes time to debug. As I am independent researching working on this project I have a limited time which I can dedicate to the project and currently do not have other team members to allocate to this task. It is for these reasons I am requesting a 3-year timeframe to conduct this project.
The UK BioBank database also has many additional factors such as lifestyle and genetics, which previous age prediction experiments did not have. So, by using these additional variables we will be able to determine how they affect neurodegeneration. In addition to improved age prediction accuracy, we will also glean insights into how these lifestyle and genetic factors change affect the neurodegeneration process.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/age-related-changes-in-dna-methylation-patterns-to-predict-drug-targets-in-osteoarthritis

Age-related changes in DNA methylation patterns to predict drug targets in osteoarthritis

Last updated:
ID:
105442
Start date:
4 October 2023
Project status:
Current
Principal investigator:
Dr Kyle Tretina
Lead institution:
Insilico Medicine AI Limited, United Arab Emirates

Around a third of the patients that receive knee replacement still have chronic pain after surgical knee replacement. We have evidence that this may be due to changes to their DNA that could be effective targets for drug development. We plan on comparing the DNA of patients with and without pain to identify the parts of the DNA that could lead us to the most important and relevant changes and lead us to an eventual treatment for those patients that have chronic pain.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/age-related-changes-in-microbiota-gut-brain-axis-with-aging-in-participants-with-different-cognitive-profiles-and-live-experiences

Age-related changes in microbiota-gut-brain axis with aging in participants with different cognitive profiles and live experiences.

Last updated:
ID:
72781
Start date:
11 October 2021
Project status:
Current
Principal investigator:
Dr Vasily Vakorin
Lead institution:
Simon Fraser University, Canada

People often make their decisions trusting their gut feelings. There is a physiological basis for that. Not only do the gut bacteria and the brain talk to each other, but they also influence each other. Gut microbes affect our mood and behaviour. In turn, the brain can change bacterial composition and function. Such communication between two fundamental systems, which goes in both directions, is known as the gut-microbiota-brain axis. Every day scientific literature offers evidence that disturbances in the relationships between the microbes and the brain have implications for a broad spectrum of health issues. These health conditions include not only various food sensitivities and gut disorders but also psychiatric disorders like depression and brain disorders like autism, Alzheimer’s and Parkinson’s disease. Understanding the influence of the gut-microbiota-brain axis on brain function and behaviour is at the forefront of neuroscience research, and this axis is essential to normal and healthy brain development.

We aim to explore how the relationships between gut microbes and the brain change in ageing and vary across individuals with different cognitive abilities and life experiences. Asking questions on the gut-microbiota-brain axis and its relation to cognition and individual life experience requires highly diverse data from many research fields. The UK Biobank provides a unique opportunity for researchers to access such data. We will look into single-nucleotide polymorphism or SNP, representing variations in single nucleotides at a specific position in the human genome, which previous studies associated with gut microbes. The brain will be described in terms of its functional networks identified by functional MRI. We will consider a wide range of cognitive tests to cluster all the participants into sub-groups, based on the similarity of participants’ cognitive profiles or life experiences such as employment history. The project is extensive in methods and relies on diverse expertise in genomics, neuroimaging, and data science. We expect to get concluding results within two years.

The impact on public health will be reached by identifying bacteria families and functional networks, which are associated with prolonged healthy cognition or risks related to individual life experiences. As we will consider an entire spectrum of ageing trajectories in the gut-microbiota-brain axis, we ultimately target the personalized recommendations for staying within those trajectories that are beneficial for a given sub-group of individuals: promoting the effects of particular bacteria with individual diets or rewiring specific brain networks with personalized rehabilitation.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/age-related-hearing-loss-brain-health-and-genetic-risk-for-alzheimers

Age-related hearing loss, brain health and genetic risk for Alzheimer’s

Last updated:
ID:
60021
Start date:
2 April 2020
Project status:
Current
Principal investigator:
Dr Judy Pa
Lead institution:
University of California, San Diego, United States of America

Age-related hearing loss is the third most common chronic health condition affecting adults in the United States, following hypertension and arthritis and has recently been identified as a risk factor for cognitive decline and dementia. Despite its high prevalence within older adults and its association with dementia, age-related hearing loss remains largely underexplored and unaccounted for in the neuroscience of aging literature. Thus, it is unclear how hearing loss may contribute to brain aging. With this study, we aim to examine how hearing loss impacts brain structure and brain function, and how the APOE-e4 gene (a known risk factor for developing Alzheimer’s disease) may influence these relationships. Our findings could point to novel intervention targets to combat brain aging and Alzheimer’s disease, including the use of hearing aids, which may provide neuroprotective effects against hearing loss.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/aging-biomarkers-better-to-prevent-disease-than-cure

Aging biomarkers – better to prevent disease than cure

Last updated:
ID:
241816
Start date:
26 November 2024
Project status:
Current
Principal investigator:
Dr Diana Domanska
Lead institution:
Oslo University Hospital, Norway

Our long term goal is to promote healthy ageing in the general population based on new knowledge from individual ageing drivers. Our short term aims are: 1) Characterise DDR ageing signatures and 2) Validate the method in a big cohort of patients from the external databases We hypothesise that individual ageing trajectories can be used to tailor healthy ageing interventions. We strongly believe that implementing effective strategies to reduce disease burden among the elderly will significantly alleviate the strain on the healthcare system. Developing of affordable therapies that promote long-term health will thereby have a huge positive impact on society. the project duration will be 36 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/aging-study-by-genotype-and-dna-methylation-in-neural-networks

Aging Study by Genotype and DNA Methylation in Neural Networks

Last updated:
ID:
56317
Start date:
1 April 2020
Project status:
Closed
Principal investigator:
Dr Yu Zhang
Lead institution:
Trinity University, United States of America

Research on predicting human biological aging has been booming during the last few decades. While traditional formula of aging prediction is based on linear models, relatively few works have explored the effectiveness of neural network models, which tends to have the advantage of learning more complex relationship from the data.

In this project, we will study three age-related diseases: cardiovascular disease, cancer, and diabetes. The duration is three years.

Neural networks are computing systems that are inspired by, but not identical to, biological neural networks that constitute human brains. A neural network learns to perform tasks by considering examples with labeled features. For example, in image recognition, they might learn to identify images that contain cats by analyzing example images that have been manually labeled as “cat” or “no cat” and using the results to identify cats in other images. They do this without any prior knowledge of cats because the neural network can learn the characteristics of cat (fur, tails, whiskers, etc.) from a large number of examples.
However, biological gnome data usually consists of hundreds of thousands of features (i.e. genetic markers), while the samples (i.e. patients) are limited due to the convoluted, expensive procedure of gathering data. This introduces the problem of overfitting which leads to poor generalization when applied to different datasets. Overfitting means that the learning is too closely or exactly to a particular set of data, and may therefore fail to fit additional data or predict future observations reliably.

In this project, we will explore several neural network models for handling biological gnome data while taking care of overfitting problems. We propose several models: Basic Neural Network, Dropout Neural Network, Least Absolute Shrinkage and Selection Operator (LASSO) Neural Network, Elastic Net Neural Network, and Correlation Pre-Filtered Neural Network (CPFNN). Our goal is to choose a model with best age prediction by comparing these models and use that model to achieve a decent result on predicting the probability of age-related disease.

The project will have several significant contributions: 1) we will develop a new direction on how to approach high dimension low sample size data in the computer science field; 2) our model can be extended in other fields which also has high dimension low sample size data such as finance; 3) we will provide a sophisticated model on age prediction and age-related disease prediction, which will have a huge impact on clinic fields.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/ai-aided-cardiovascular-outcomes-prediction-using-cardiac-imaging-biomarkers

AI-aided Cardiovascular outcomes prediction using cardiac imaging biomarkers

Last updated:
ID:
72280
Start date:
20 June 2023
Project status:
Current
Principal investigator:
Anant Madabhushi
Lead institution:
Emory University, United States of America

Cardiovascular diseases are among the leading causes of mortality across the world. In particular, coronary artery disease represent a high burden of cardiovascular deaths. The aims of our study is to develop artificial intelligence-based tools using MRI scans, EKG, and patient demographics to predict the risk of adverse outcomes in patients

Type of dataset required: Cardiac imaging data (CMR), patient demographics, outcomes, QOLs, EKGs.

The expected value of research (public interest) is that the development of a non-invasive, novel, AI-aided toolkit will allow accurate prediction of the risk of adverse cardiovascular outcomes. This toolkit will help in future prospective studies to preemptively identify high-risk patients for whom protective strategies may be targeted.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/ai-and-iot-based-multi-disease-prediction-framework-for-geriatric-healthcare

AI and IoT-Based Multi-Disease Prediction Framework for Geriatric Healthcare

Last updated:
ID:
936124
Start date:
18 September 2025
Project status:
Current
Principal investigator:
Mrs Anjana Prasad
Lead institution:
Birla Institute of Technology and Science (BITS), Pilani, India

This research aims to develop an AI driven multi disease framework for early detection and risk stratification of chronic diseases common in the elderly such as diabetes, cardiovascular diseases, and neurodegenerative conditions by integrating data from multimodal healthcare sources. By leveraging large-scale biomedical data from the UK Biobank, the research will help identify high-risk individuals and support precision medicine strategies that can improve health outcomes and reduce long-term healthcare costs.

Scientific Rationale:

Chronic diseases in elderly individuals often co-occur, and existing diagnostic pathways are inadequate in addressing comorbidities holistically. Recent advancements in machine learning and IoT-based sensing provide an opportunity to develop predictive models that integrate multi-modal data. The UK Biobank offers a unique, richly annotated dataset ideal for training and validating these models due to its extensive genotypic, phenotypic, and imaging data.

Aims and Objectives:

1. Develop an AI driven healthcare framework for comprehensive prediction of multiple chronic disorders in elderly patients and model their progression over time.
2. Identify key biomarkers and risk patterns to support early intervention and monitoring by integrating genomic, biochemical, lifestyle, and imaging data to enhance prediction accuracy.
3. Optimize AI model for real time deployment and develop a personalized health management system.
4. Compare the model performance against traditional methods and validate the AI-IoT framework to assess its clinical impact.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/ai-based-analysis-of-taste-changes-and-their-associations-with-oral-health-systemic-diseases-and-genetic-or-protein-biomarkers-using-uk-biobank

AI-based Analysis of Taste Changes and Their Associations with Oral Health, Systemic Diseases, and Genetic or Protein Biomarkers Using UK Biobank

Last updated:
ID:
832838
Start date:
23 July 2025
Project status:
Current
Principal investigator:
Mr ZhiHao Zhang
Lead institution:
Yonsei University, Korea (South)

Taste alterations, preferences, and disorders are emerging as early indicators of both systemic and oral health conditions. However, their mechanisms remain poorly defined and difficult to quantify. This project leverages the UK Biobank’s large-scale, multi-modal dataset to examine the relationships between taste changes, oral health, systemic diseases, and dental treatments. It also explores genetic, proteomic, and biomarker factors involved in taste dysfunction using advanced machine learning.
Research Questions:
1. How are taste changes (loss, distortion, sensitivity) associated with oral health factors like periodontal disease, tooth loss, and prosthesis use?
2. What links exist between taste alterations and systemic diseases, including metabolic, cardiovascular, and neurodegenerative conditions?
3. Which genetic variants and protein biomarkers are associated with taste dysfunction or altered taste preferences?
4. Can deep learning models quantify a “Taste Alteration Risk Score” based on integrated clinical and molecular data?
Objectives:
1. Analyze the distribution of taste changes using ensemble models (e.g., XGBoost, LightGBM).
2. Identify oral and systemic factors associated with taste dysfunction via multivariate models and explainable AI (e.g., SHAP).
3. Discover genetic and proteomic features using dimensionality reduction, autoencoders, and CNNs on GWAS/proteomic data.
4. Build predictive models to: (a) forecast taste dysfunction from health profiles, and (b) predict disease risk from taste changes using GNNs and survival analysis.
The rationale is that taste disturbances reflect interactions between oral/systemic health and molecular mechanisms. By integrating machine learning with UK Biobank data, this study aims to objectively quantify taste dysfunction, inform early disease detection, identify biomarkers and targets, and guide innovation in dental and medical care.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/ai-based-assistance-for-the-diagnosis-of-atrial-fibrillation

AI-based assistance for the diagnosis of atrial fibrillation

Last updated:
ID:
197590
Start date:
12 December 2024
Project status:
Current
Principal investigator:
Professor Britta Wrede
Lead institution:
Bielefeld University, Germany

Our research aims to leverage machine learning methods to assist in the diagnosis of Atrial Fibrillation. Atrial Fibrillation is one of the most common arrhythmias and is associated with a higher risk of stroke. It occurs in episodes and when active, can be identified by its abnormal rhythm on an electrocardiogram (ECG). However, often there is no active episode, when the patient is at the doctor’s office and diagnosis requires long-term monitoring with portable ECG machines at home. Using machine learning to identify the disease, even when the current rhythm of the heart appears as healthy on the ECG, could lead to reduced time to diagnosis and therefore, ease the burden and risk of stroke in patients.
During the three year duration of the project we plan to research different machine learning methods and find the best algorithms for processing ECG data. Furthermore we plan to research different means to explain the results from the machine learning model to the clinical personal and patients making the results reliable and trustworthy. In order to achieve this, we plan to make use of the inherent geometrical structure of the signals from the different ECG electrodes, as well as expert knowledge from doctors.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/ai-based-detection-of-early-signs-of-alzheimers-disease-using-retinal-fundus-images

AI-based Detection of Early Signs of Alzheimer’s Disease Using Retinal Fundus Images

Last updated:
ID:
915007
Start date:
19 September 2025
Project status:
Current
Principal investigator:
Mrs Kamily Eduarda da Silva
Lead institution:
Fundação Escola Tecnica Liberato Salzano Vieira da Cunha, Brazil

Alzheimer’s disease is a neurodegenerative disorder with a long preclinical stage. Early detection is crucial for timely intervention and slowing disease progression. Recent research suggests that subtle retinal changes may be early indicators of neurodegenerative processes, offering a non-invasive diagnostic opportunity.
This project aims to develop an AI-based diagnostic tool capable of detecting early signs of Alzheimer’s disease through analysis of retinal fundus images. We will use machine learning and deep learning techniques to identify patterns associated with early cognitive decline. The research will include training and validating models using retinal images and associated cognitive test data from the UK Biobank.
Our objectives include:
– Identifying imaging biomarkers of Alzheimer’s in retinal fundus photos
– Developing an AI model with high sensitivity and specificity
– Evaluating the model’s performance against established clinical variables
– Contributing to early detection methods in a scalable and non-invasive way
The project is aligned with public interest and health improvement goals, supporting the early diagnosis of a globally prevalent and burdensome disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/ai-based-diagnosis-prediction-and-precision-medicine-of-metabolic-diseases

AI-based diagnosis, prediction and precision medicine of metabolic diseases

Last updated:
ID:
76457
Start date:
9 February 2022
Project status:
Current
Principal investigator:
Professor Xiao-Hua Zhou Zhou
Lead institution:
Peking University, China

In this project, we aim to address the following issues mainly in the area of liver diseases, e.g., non-alcoholic fatty liver disease (NAFLD), nonalcoholic steatohepatitis (NASH), cirrhosis, liver cancer.
1. Find tools/indicators to identify who is more likely to develop NAFLD from obese or to develop NASH from NAFLD and etc.;
2. Find tools/indicators in assisting the clinical diagnosis of NAFLD, NASH and etc.;
3. Find tools/indicators to identify stages of NAFLD, NASH and etc.;
4. Find the best treatments for different individuals with liver diseases;
5. Infer the impact of medical history (e.g., NAFLD, NASH) on the prognosis and endpoints of COVID-19 infections.
Current NAFLD diagnosis relies on highly invasive and risky liver biopsies which cannot be applied to all patients at risk, and in addition, it cannot predict disease progression and remains unknown which patients develop NAFLD secondary to obesity. Hence, there is a need to identify tools/indictors which can better identify the disease, who should be targeted for interventions, what the risks are and differential response to therapy response and etc.
Whole body MRI facilitates the visualization of total body fat and lean mass distribution, which is associated with the risk of obesity related diseases and abnormal metabolic processes, while retina fundus (eye) images have been used to diagnose and predict disease (e.g. Type 2 diabetes, Cardiovascular diseases, Major Adverse Cardiovascular Events). In addition, many of the classical indictors for liver dysfunction are known to be in the category of plasma proteins. Hence, we envision to apply statistical and machine learning approaches to gain knowledge in MRI, retina fundus images and proteomics to improve the diagnosis and prediction of liver diseases.
To achieve these aims, we want to access annotated image, gene and proteomics data from the UK Biobank. We believe that these high dimensional data in UK biobank has the potential to improve the current diagnosis, prediction and treatment selection methods to liver diseases.
This project is an on-going project and the first stage would be 3 years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/ai-based-integration-of-multi-omic-data-for-precision-medicine-in-movement-disorders

AI based integration of multi omic data for precision medicine in movement disorders

Last updated:
ID:
72354
Start date:
11 November 2022
Project status:
Current
Principal investigator:
Dr Thomas Welton
Lead institution:
National Neuroscience Institute, Singapore

Essential tremor and Parkinson’s disease are common movement disorders. They are difficult to diagnose and monitor because there is no single factor that tells us their status, and there are many intermixing factors which cause them. This project aims to create new ways of investigating the causes of movement disorders using artificial intelligence (AI) to combine data across different levels of biology, from the genetic, to markers present in the blood, to brain structure and behaviour. The AI will be able to identify combinations that are relevant for clinical status. This could provide a clearer understanding of the causes of these diseases, which will allow doctors to diagnose earlier and be more informed in their treatment decisions.
Our group has access to large Parkinson’s and essential tremor cohorts with high quality magnetic resonance imaging (MRI) and other biomarker data. We aim to identify new signatures of these disorders, to understand how combined information from multiple biological scales might help in diagnosis and subtyping, and to test for replication and validation of these findings in a large combined sample which will include the UK Biobank data.
We are requesting 36 months’ access to data from subjects with movement disorders and controls who had brain MRI (MRI, demographic, clinical, cognitive, behavioural, genetic and biomarker data).


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/ai-based-integrative-risk-scores-for-predictive-medicine-and-precision-therapy-in-complex-and-rare-diseases

AI-based integrative risk scores for predictive medicine and precision therapy in complex and rare diseases

Last updated:
ID:
96802
Start date:
19 January 2023
Project status:
Current
Principal investigator:
Dr Stavroula Kanoni
Lead institution:
Queen Mary University of London, Great Britain

The evolution of the artificial intelligence (AI) is promising significant advances in the area of health science and health care. With our ability to harvest a significant amount of information from large populations, like UK Biobank, we are extending our horizons on better prevention, diagnosis and treatment of diseases. AI methods would allow us to better process and combine multiple layers of health-related information, including genetic predisposition, gene expression, clinical and biochemical characteristics, lifestyle and behavioral factors, in order to customize the way we predict the disease risk at an individual level, personalize the prevention strategies and offer tailored treatment options that maximize the therapeutic effect. We hypothesize that by using sophisticated analytical methods and a range of potential predictors for a disease, we can create novel, high-accuracy prediction tools that could be easily implemented in the clinical practice. To that aim, we propose to utilize the deep phenotyping and multi-modal measurements of the UK Biobank resource to develop, test and fine-tune these AI-based integrated risk assessment tools. We aspire that the adoption of such novel integrated risk scores in clinical practice, will be beneficial for reducing NHS costs, both due to more effective disease prevention and disease management. To comprehensively assess how such tools might be accurate across different diseases, we have selected to investigate a range of high burden diseases, including the cardiovascular disease spectrum, type 2 diabetes, obesity, familial hypercholesterolemia, fatty liver disease, thyroid disease, frailty and COVID-19. We will be focusing both on common and rare sub-phenotypes of these diseases, to further fine-tune the utility of our tools for patient care. We plan to validate our novel tools to other population cohorts and assure transferability to non-European ancestry groups. Finally, we plan to test these AI-based integrated risk scores in clinical trial settings. We anticipate our project to last for 3 years at minimum.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/ai-based-long-term-health-risk-evaluation-for-driving-behaviour-change-strategies-in-children-and-youth-smartchange

AI-based long-term health risk evaluation for driving behaviour change strategies in children and youth – SmartCHANGE”

Last updated:
ID:
194358
Start date:
18 March 2025
Project status:
Current
Principal investigator:
Professor Mitja Lustrek
Lead institution:
Jozef Stefan Institute, Slovenia

Non-communicable diseases (NCDs) are the leading cause of death and healthcare expense. Common risk factors for many of them are obesity and low physical fitness resulting from an unhealthy lifestyle. Targeting children and youth for lifestyle interventions has been suggested because early precursors of most NCDs are already present at this age, childhood and adolescence are critical periods for the acquisition of healthy lifestyle habits, and unhealthy lifestyle in this age group is prevalent.
We propose to develop long-term risk-prediction models for cardiovascular and metabolic disease for people aged 5-19. We have already identified 15 datasets with data on behaviour, fitness, biomarkers and actual NCDs spanning various ages. We will develop machine-learning methods that can train models on such heterogeneous datasets, enabling the prediction of risk for people of various ages for whom different data is available. We will employ federated learning for data privacy, carefully curate and balance the data to ensure it is bias-free and representative of the target group, and employ methods for explanation and visualisation of the data, models and predictions.
Cardiovascular and metabolic health trajectories established during childhood and adolescence play a crucial role in shaping an individual’s well-being throughout their life. While existing cohort studies provide valuable insights into early-life risk factors, the absence of data extending into older ages poses a significant limitation. Recognizing this gap, the inclusion of the UK Biobank datasets becomes imperative for a comprehensive understanding of the lifelong impact of childhood predictors on cardiovascular and metabolic outcomes.
The UK Biobank, with its extensive dataset covering a wide age range, offers a unique opportunity to bridge the temporal gap in existing cohort studies.
Including the UK Biobank datasets enriches our predictive models by providing a more diverse and extensive set of variables. This augmentation allows for a refined analysis of the relationships between childhood predictors (such as lipid profiles, physical activity, and physical fitness) and later cardiovascular and metabolic events. The larger sample size and varied demographic characteristics within the UK Biobank contribute to the robustness and generalizability of our predictive models, with far-reaching implications for individual well-being and public health, positioning our project at the forefront of unraveling the intricate connections between early-life factors and lifelong cardiovascular and metabolic health.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/ai-driven-analysis-of-fundus-camera-images-for-cardiovascular-diseases-detection

AI-Driven Analysis of Fundus Camera Images for Cardiovascular Diseases Detection

Last updated:
ID:
240599
Start date:
3 October 2024
Project status:
Current
Principal investigator:
Mr Jan Bayer
Lead institution:
Aireen a.s., Czechia

Our project aims to use advanced computer technology to study how images of the eye can help detect early signs of heart problems. By analyzing special pictures taken with a fundus camera, we hope to find subtle clues that could indicate issues with the heart. These clues might be things like tiny changes or patterns in the eye that can’t be seen with the naked eye. Our project will take some time, likely a few years, as we carefully analyze the data and develop new tools to help doctors identify these eye changes. If successful, our research could lead to better ways of spotting heart problems early, potentially saving lives by allowing doctors to intervene sooner. This could have a significant impact on public health by improving heart disease prevention and treatment strategies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/ai-driven-closed-loop-system-for-transcutaneous-vagus-nerve-stimulation-in-glucose-control

AI-Driven Closed-Loop System for Transcutaneous Vagus Nerve Stimulation in Glucose Control

Last updated:
ID:
577799
Start date:
21 April 2025
Project status:
Current
Principal investigator:
Mr Zijia Wang
Lead institution:
Imperial College London, Great Britain

The global rise in metabolic disorders, such as type 2 diabetes (T2D) and obesity, demands innovative, non-invasive therapies. This project aims to develop an AI-driven closed-loop system using transcutaneous vagus nerve stimulation (tVNS) to regulate glucose metabolism via GLP-1 (glucagon-like peptide-1) modulation. By leveraging machine learning, the system dynamically adjusts tVNS parameters to optimize glucose levels.

Key objectives include:

Modeling the tVNS-GLP-1-Glucose Relationship: Developing AI models to map the pathways between tVNS, GLP-1 secretion, and glucose regulation.
Designing Adaptive Algorithms: Using advanced AI techniques to personalize tVNS stimulation for precise glucose control.
Exploring Lifestyle and Genetic Factors: Incorporating UK Biobank data to refine predictions and enable personalized therapies.
Validating Efficacy: Simulating and evaluating the system using UK Biobank data as an alternative to pharmacological treatments.
In compliance with UK Biobank’s AI guidelines, findings will be disseminated via peer-reviewed publications and conferences, ensuring adherence to ethical standards and confidentiality. Derived variables will be returned to UK Biobank, and AI models will be developed following best practices, emphasizing safety, transparency, and fairness. This project leverages UK Biobank’s extensive datasets to create scalable, personalized, and non-invasive glucose control solutions, advancing public health.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/ai-driven-early-screening-progression-prediction-and-biomarker-identification-for-cardiovascular-and-neuropsychiatric-diseases

AI-Driven Early Screening, Progression Prediction, and Biomarker Identification for Cardiovascular and Neuropsychiatric Diseases

Last updated:
ID:
681047
Start date:
26 June 2025
Project status:
Current
Principal investigator:
Professor Zhengxing Huang
Lead institution:
Zhejiang University, China

This research aims to explore the progression, early detection, and biomarker identification of cardiovascular and neuropsychiatric diseases, including heart failure, cardiomyopathy, atrial fibrillation, myocardial infarction, and Alzheimer’s disease. By leveraging clinical and imaging data, the key objectives are:
Predict Disease Progression: To develop AI-driven models for predicting the progression of these diseases, aiding in better forecasting of patient outcomes and guiding clinical decision-making.
Identify Biomarkers: To identify biomarkers that reflect the early stages or progression of these diseases, enabling earlier diagnosis and more personalized treatment strategies.
Early Screening: To design screening tools that detect high-risk individuals or early-stage diseases, facilitating timely intervention and improving patient prognosis.
Examine Disease Interactions: To investigate how cardiovascular and neuropsychiatric diseases, along with their comorbidities, interact and influence each other, potentially guiding more effective treatment approaches.
Scientific Rationale:
Cardiovascular and neuropsychiatric diseases are major contributors to global morbidity and mortality, with complex progression patterns that are often difficult to predict. Early detection and personalized treatment strategies are key to improving patient outcomes. Clinical and imaging data, combined with AI, offer significant potential to uncover hidden patterns in disease progression, identify early biomarkers, and enable more timely interventions. AI models can automate the analysis of multidimensional data, improving prediction accuracy and facilitating early diagnosis. Understanding disease interactions, particularly with comorbidities, is also critical for optimizing treatment. This research aims to leverage AI to advance early detection, disease progression prediction, and biomarker identification, ultimately improving clinical care and disease management.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/ai-driven-identification-of-early-imaging-biomarkers-for-predicting-multi-organ-aging-and-frailty-using-whole-body-mri-and-pet

AI-Driven Identification of Early Imaging Biomarkers for Predicting Multi-Organ Aging and Frailty Using Whole Body MRI and PET

Last updated:
ID:
105529
Start date:
1 September 2023
Project status:
Current
Principal investigator:
Dr Keno Bressem
Lead institution:
Charite - Universitatsmedizin Berlin, Germany

In this three-year research project, we will investigate a novel method for identifying people at risk of developing cancer or heart disease using artificial intelligence (AI). Our goal is to see if the age of a person’s organs, as determined by MRI scans, can act as an early warning sign for these conditions. The main idea is that unhealthy organs age more quickly, and by studying organ age, doctors might predict disease likelihood and take preventive action.
The basis for this project is the understanding that aging affects people and their organs differently. By looking at organ age, we hope to uncover the potential of this new marker for predicting disease risk and guiding personalised treatments.

During the project, we’ll first teach an AI model to identify and separate organs in MRI images. Next, using a group of patients without major diseases, the AI model will learn to predict the age of individual organs. The model will then be used on people with known major health problems, like cancer and heart disease, to estimate the biological age of their organs.

Our project will also develop AI models for predicting frailty, sarcopenia, and osteoporosis. These models will consider factors like muscle mass, bone density, and functional performance measures. This will enable a more comprehensive assessment of an individual’s overall health and risk for age-related conditions.

By tapping into the vast UK Biobank dataset, this research could revolutionise preventive strategies and public health efforts by enabling early detection of organs at risk and other surrogate biomarkers, ideally before disease onset. The findings may lead to targeted prevention measures, reducing healthcare system strain and improving the quality of life for an aging population. Furthermore, the study’s results could significantly influence public health policies, promoting healthy lifestyles and reducing risks linked to age-related conditions.

In conclusion, this three-year project aims to use AI technology to explore organ age as a potential early sign of cancer and heart disease risk. If successful, this novel approach could lay the foundation for more effective prevention strategies and better public health outcomes, ultimately benefiting people in the UK and around the world.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/ai-driven-integration-of-multi-modal-data-for-biomarker-discovery-in-early-mental-disorder-diagnosis-and-prediction

AI-Driven Integration of Multi-Modal Data for Biomarker Discovery in Early Mental Disorder Diagnosis and Prediction

Last updated:
ID:
581377
Start date:
28 August 2025
Project status:
Current
Principal investigator:
Dr Ji-Won Chun
Lead institution:
Catholic Medical Center (Korea), Korea (South)

This research aims to identify biomarkers from multi-modal data sources, including genetic information, neuroimaging, cognitive assessments, lifelog data, and histories of substance use, such as alcohol or drugs. By analyzing these diverse data types, the study seeks to uncover biological, cognitive, and behavioral indicators associated with mental health conditions, including the effects of addiction on mental well-being.
It also explores how deep learning and AI techniques can process multi-modal data to enhance classification models for diagnosing mental health disorders. The focus is on improving model accuracy and identifying patterns across complex data sources, including the impact of external environmental factors on mental and physical health.
The integration of multi-modal data is critical for developing explainable AI models that provide transparent and interpretable insights for mental health diagnosis and prediction. Identifying effective methods for data fusion and feature selection is essential to ensure these models are both accurate and understandable.
Finally, this research examines the validation and optimization of AI models for use in digital healthcare devices, enabling real-time diagnosis and prediction of mental health conditions. The study also investigates how external environmental factors, such as stressors and lifestyle influences, contribute to mental health outcomes, ensuring these models are robust, reliable, and applicable across diverse populations.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/ai-driven-multimodal-biomarkers-for-early-diagnosis-of-alzheimers-disease

AI-Driven Multimodal Biomarkers for Early Diagnosis of Alzheimer’s Disease

Last updated:
ID:
740106
Start date:
21 May 2025
Project status:
Current
Principal investigator:
Dr Farshid Hajati
Lead institution:
University of New England, Australia

Research Questions:
1. Can machine learning (ML) models diagnose Alzheimer’s disease (AD) using multimodal UK Biobank data, including clinical, cognitive, lifestyle, demographic, and imaging variables?
2. What are the most significant risk factors and biomarkers associated with AD progression, and how do they interact?
3. How does integrating retinal imaging with other health-related variables improve predictive accuracy for early AD diagnosis?
4. How can domain adaptation enhance the generalizability of AD diagnosis models across diverse populations?
5. Can feature importance mapping provide interpretable insights to support clinical decision-making in AD diagnosis?

Objectives:
* Develop and validate ML models for AD diagnosis using UK Biobank’s multimodal dataset.
* Identify key demographic, medical, cognitive, and imaging biomarkers contributing most to AD diagnosis.
* Assess the added value of retinal imaging when combined with other biomarkers for AD risk prediction.
* Apply domain adaptation techniques to improve model robustness and ensure generalizability, minimizing dataset bias.
* Utilize explainability techniques (e.g., SHAP, Grad-CAM) to interpret model diagnosis and enhance clinical applicability.

Scientific Rationale:
Alzheimer’s disease is a progressive neurodegenerative condition with no cure, making early diagnosis essential for intervention. However, traditional diagnostic methods, such as neuroimaging and cerebrospinal fluid analysis, are invasive, costly, and often performed late. Machine learning models trained on large-scale datasets, such as UK Biobank, offer an opportunity to develop scalable, non-invasive screening tools. A major challenge in AI-driven healthcare is ensuring model generalizability across populations. Domain adaptation techniques will mitigate distributional shifts between training and target populations.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/ai-driven-prediction-of-cardiovascular-disease-using-electrocardiogram-genomics-proteomics-and-imaging-biomarkers

AI-Driven Prediction of Cardiovascular Disease Using Electrocardiogram, Genomics, Proteomics, and Imaging Biomarkers

Last updated:
ID:
836794
Start date:
8 August 2025
Project status:
Current
Principal investigator:
Dr Lu Liu
Lead institution:
Dalian University, China

This project aims to develop advanced artificial intelligence (AI) models to accurately predict cardiovascular disease (CVD) risk using multimodal data-electrocardiogram (ECG), genomics, proteomics, and imaging biomarkers-from UK Biobank. CVD is the leading global cause of morbidity and mortality, yet traditional risk models lack precision. Integrating AI with rich biomedical data offers the potential for earlier, personalized intervention.

Research questions include:

How can AI models detect early ECG-based signals predictive of future cardiovascular events?

Which genomic and proteomic features significantly improve risk prediction when combined with clinical profiles?

Can machine learning enhance the interpretation of cardiac imaging (MRI, CT) for CVD detection and prognosis?

How can multimodal data integration via AI provide a comprehensive cardiovascular risk stratification?

Objectives:

Develop deep learning algorithms for ECG-based risk prediction.

Identify key genetic and proteomic biomarkers for enhanced model performance.

Apply ML techniques to imaging-derived cardiac phenotypes.

Build explainable, integrative AI models across all modalities for clinical deployment.

Dissemination and compliance with UK Biobank’s AI policy:

Models will be developed and validated with interpretability in mind, using explainable AI techniques (e.g., SHAP, saliency maps). We will publish all findings in peer-reviewed open-access journals, and make AI model code, trained weights, and relevant scripts available on GitHub or institutional repositories in accordance with UK Biobank’s AI policy. The outputs will be presented at public scientific meetings to maximize transparency and utility.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/ai-driven-radiogenomic-and-body-composition-analysis-for-abdominal-tumor-diagnosis-and-prognosis

AI-Driven Radiogenomic and Body Composition Analysis for Abdominal Tumor Diagnosis and Prognosis

Last updated:
ID:
940419
Start date:
29 July 2025
Project status:
Current
Principal investigator:
Professor Lian Yang
Lead institution:
Union Hospital, Tongji Medical College, China

1. Research Questions & Objectives
Abdominal tumors (e.g., hepatocellular carcinoma) cause high mortality worldwide. Non-invasive stratification of tumor biology, body composition, and immune status using imaging, multi-omics, and clinical data remains challenging and benefits from large cohorts like UK!Biobank.
1.1 Primary Question
Can an AI-driven radiomics signature from abdominal CT/MRI-integrated with genetics, proteomics, metabolomics, body composition, lifestyle, and clinical data-detect and predict prognosis of abdominal tumors more effectively than standard clinical measures?
1.3 Objectives
1.Build and validate an AI radiomics pipeline to extract CT/MRI features.
2.Link imaging features with germline genetics (e.g., polygenic risk).
3.Combine imaging with proteomic, metabolomic, and clinical markers for diagnostic/prognostic evaluation.
4.Analyze body composition (VAT/SAT, muscle index) in relation to tumor phenotype and outcomes.
5.Assess lifestyle and medical history effects on radiogenomic signatures.
6.Develop AI models for tumor detection, treatment response, and survival prediction.
7.Compare multimodal models against clinical-only models to measure added value.
2. Scientific Rationale
Radiomics quantifies imaging features from CT/MRI to detect tumor heterogeneity beyond visual assessment, improving HCC grading, staging, and prognosis. Radiogenomics demonstrates imaging signatures can non-invasively predict genetic, transcriptomic, proteomic, metabolomic, and immune characteristics. Body composition metrics-like visceral fat and muscle mass-reflect metabolic health and inflammation, influencing tumor progression. The UK!Biobank provides extensive imaging, multi-omics, lifestyle, and outcome data, ideal for AI-driven integrative research. Our approach promises to improve non-invasive tumor profiling, support personalized treatment, reduce invasive biopsies, and advance precision oncology.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/ai-driven-retinal-analysis-for-early-detection-of-neurocognitive-disorders-and-modifiable-risk-factors

AI-Driven Retinal Analysis for Early Detection of Neurocognitive Disorders and Modifiable Risk Factors

Last updated:
ID:
775744
Start date:
28 April 2025
Project status:
Current
Principal investigator:
Dr Daniel Raff
Lead institution:
Florence Biosciences Inc., Canada

Rationale
Retinal imaging non-invasively captures microvascular and neurological features associated with neurocognitive disorders such as Alzheimer’s disease and vascular dementia, as well as systemic conditions like dyslipidemia or hypertension, which are established modifiable risk factors for cognitive decline. Optical coherence tomography (OCT) and color fundus photography (CFP) are widely deployed modalities for monitoring ocular health. Their broad clinical adoption and widespread availability position them as viable tools for early disease detection. By leveraging large unannotated datasets for pretraining, followed by fine-tuning with smaller labeled datasets, vision transformer models coupled with multi-task learning (MTL) can generalize well to unseen data, making them ideal candidates for analyzing retinal images in the context of neurocognitive decline.

Research Question
Based on this context, we propose the following research question: Can retinal images combined with genetic and clinical data, accurately predict the risk of neurocognitive disorders?

Objectives
Our objective is to integrate retinal imaging data, genetic information, and clinical outcomes, to provide a robust, non-invasive tool to assess cognitive impairment. This MTL system will primarily assess neurocognitive disease risk but is hypothesized to perform optimally when concurrently trained to detect modifiable risk factors and monogenic variants associated with these risk factors and neurocognitive disorders.

A 2024 Lancet report (Livingston) indicated that modifying risk factors may prevent or delay nearly half of dementia cases, and as such there is value in presenting individuals with both their neurocognitive disorder risk, alongside their risk of established modifiable risk factors to motivate change. Developing a non-invasive, accessible tool for early risk stratification may empower individuals to proactively manage and reduce their risk of neurocognitive disorders.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/ai-driven-risk-profiling-of-amyotrophic-lateral-sclerosis-als-patient-subgroups-based-on-blood-and-omics-biomarkers-in

AI-Driven risk profiling of Amyotrophic Lateral Sclerosis (ALS) patient subgroups based on blood and omics biomarkers in

Last updated:
ID:
348061
Start date:
21 January 2025
Project status:
Current
Principal investigator:
Mr Tim Mark Chambers
Lead institution:
Accenture Global Solutions Ltd., Ireland

Amyotrophic Lateral Sclerosis (ALS) is a progressive neurodegenerative disease that affects motor neurons, leading to muscle weakness and eventual paralysis. Most patients face a grim prognosis, with 70% succumbing within three years of diagnosis. Our recent study analysed a proprietary dataset from an ALS cohort, revealing intriguing findings:

* Metabolic Clues: We observed a paradoxical association-a lipid profile linked to cardiovascular risk appeared to reduce ALS risk. Additionally, metabolic disorders were more prevalent in ALS cases than in controls.
* Geographical Considerations: To validate our results, we’ll compare them with broader datasets, including UK Biobank data. Our proprietary dataset, derived from the US population, necessitates broader geographical representation.
* Predicting ALS Progression: Time matters in ALS progression. We aim to develop machine learning methods to detect pre-symptomatic metabolic changes. Early detection could revolutionize ALS management.
* In-House and UK Biobank Data: Our focus includes ~11,500 ALS patients and a matched control group. We’ll analyse lab observations and blood biomarkers, comparing results with UK Biobank cohorts.
* Method Two: Our innovative machine learning techniques will explore temporal aspects of patient data, considering disease progression acceleration.
Impact and Benefits:
* Public Awareness: Our research will raise awareness of early ALS risk factors, empowering individuals to seek timely intervention.
* Practitioner Insights: Healthcare professionals can proactively identify at-risk patients, potentially improving outcomes.
* Scientific Advancements: Novel biomarkers may guide treatment decisions and drug development.
* Personalized Health Monitoring: High-risk individuals can monitor their metabolic profiles for early signs.
In summary, this project bridges research and practical applications, aiming to transform ALS care.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/ai-driven-understanding-of-the-brain-imaging-and-genetic-basis-of-complex-diseases-traits

AI-Driven Understanding of the Brain Imaging and Genetic Basis of Complex Diseases/Traits

Last updated:
ID:
926220
Start date:
2 October 2025
Project status:
Current
Principal investigator:
Professor Fuqing Zhou
Lead institution:
The First Affiliated Hospital of Nanchang University, China

This project aims to leverage the vast UK Biobank (UKB) dataset by integrating Artificial Intelligence (AI), multimodal brain imaging (fMRI, DTI, sMRI), clinical indicators, and Genome-Wide Association Study (GWAS) data. Our central research question is: How do genetic variations mediate the development and progression of complex diseases/traits by influencing brain structure and function? We will investigate a broad spectrum of conditions including cancers, various tumors, chronic pain, anxiety-depression disorders, aging-related traits, neurological disorders (e.g., Alzheimer’s, Parkinson’s), psychiatric disorders (e.g., schizophrenia, bipolar disorder), and cardiovascular and cerebrovascular diseases, along with their associated risk factors.
Our primary objectives are: 1) To identify novel genetic associations and their corresponding brain imaging phenotypes for these complex conditions. 2) To elucidate the causal pathways through which genetic variants influence brain structure and function, thereby contributing to disease risk and progression. 3) To develop robust AI-driven predictive models for disease incidence and patient subtyping based on integrated genetic, imaging, and clinical data. This research is scientifically rational as it addresses the persistent challenge of “missing heritability” in complex diseases, providing a comprehensive, data-driven approach to unraveling intricate gene-brain-disease relationships. By leveraging UKB’s unprecedented scale and depth, we aim to uncover previously unrecognized biological insights.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/ai-empowered-digital-twining-of-the-heart-based-on-multi-modal-data

AI-empowered Digital Twining of the heart based on Multi-Modal Data

Last updated:
ID:
1008726
Start date:
5 November 2025
Project status:
Current
Principal investigator:
Dr wenjing Xiang
Lead institution:
Southern University of Science and Technology, China

1.Research Questions:
(A)How can multi-modal, longitudinal, and cross-organ data be integrated to accurately model individual cardiac structure and function using cardiac digital twins?
(B)What genetic, biochemical, lifestyle, and environmental factors contribute to early, subclinical cardiac changes?
(C)Can personalised cardiac digital twins models offer new features to aid predictions for disease onset and progression?
2.Objectives:
(A)Develop high-precision cardiac digital twin models by combining UK Biobank imaging, biochemical, genomic/proteomic, and lifestyle/environmental data.
(B)Identify early biomarkers and phenotypic signatures associated with increased cardiovascular risk.
(C)Model dynamic interactions between cardiac and systemic health indicators to predict individual disease trajectories.
(D)Evaluate the clinical utility of simulation-based predictions for prevention and intervention strategies.
3.Scientific Rationale:
Cardiovascular disease often progresses silently until irreversible damage occurs. AI-driven digital twin technology offers a novel means to detect early changes, forecast progression, and personalise interventions. The UK Biobank’s large, standardised, multi-modal dataset-spanning imaging, omics, biochemical, lifestyle, and follow-up health outcomes-provides an unparalleled foundation for robust model development and validation. Leveraging these resources will enable building a digital twinning framework that is reproducible, generalisable, and thus offers clinically relevant findings, contributing to more effective strategies for reducing the global CVD burden.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/ai-enabled-modeling-for-biomarker-and-drug-target-identification

AI enabled modeling for biomarker and drug target identification.

Last updated:
ID:
289664
Start date:
4 February 2025
Project status:
Current
Principal investigator:
Dr Kevin Litchfield
Lead institution:
Isomorphic Labs Limited, Great Britain

Current drug development processes require on average 10-15 years to provide a new medicine, leaving a long wait for patients to experience clinical benefit. Recent developments in artificial intelligence (AI) are enabling processes to be accelerated, and this research proposal aims to confirm whether AI can speed up particular steps in the drug development process. In particular, AI can support process improvements in the pre-clinical stages of drug development, i.e., before medicines are used in humans. For example, aim (i) of this study is to use to AI to understand the biological links between genes and diseases, by processing large volumes of data rapidly. In addition, in aim (ii) we plan to use AI models to enable accelerated design of diagnostic tests, to understand if a drug is fit for purpose (biomarker tests). Previous work we have undertaken has demonstrated AI models can be useful in drug development – for example, in predicting the structure of proteins, which are the key targets that medicines act against. Traditional methods to understand protein structure can take years of experimental work in a laboratory, while AI models can make immediate predictions, hence allowing drug design work to progress more quickly. This study will use genetic, imaging and blood test data, compared with health outcomes, to develop AI models to support drug development efforts. The study will run over three years and results from this work will support the development of new medicines for chronic diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/ai-enabled-structure-and-function-analysis-for-establishing-the-eye-as-the-window-to-the-heart-and-brain

AI Enabled Structure and Function Analysis for Establishing the Eye as the Window to the Heart and Brain

Last updated:
ID:
54078
Start date:
4 February 2020
Project status:
Current
Principal investigator:
Professor Yalin Zheng
Lead institution:
University of Liverpool, Great Britain

This project aims to study whether the eyes are the windows to the heart and brain. The human eyes are transparent in nature and thus easy to be imaged without direct contact and radiation. The eyes are ideal to study problems of the other critical organs such as the brain and heart which include, but not limited to, dementia, hypertension, stroke, heart disease and so on.

In this project we will explore the unique potential of the UKBiobank imaging dataset for improving the health of future generations. We plan to apply artificial intelligence techniques for the analysis of images of the eye, heart and brain so as to generate important features to represent the health of these organs and their relationships. Based on the geometries obtained from image analysis, we will study the blood flow patterns of the eyes and physical properties of the heart by developing sophisticated computer models. The outputs from the project will help us to understand disease mechanism, prevent and diagnose critical conditions in an early manner in the future.

The project will last 36 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/ai-in-alzheimer-diagnostics

AI in Alzheimer Diagnostics

Last updated:
ID:
321004
Start date:
22 January 2025
Project status:
Current
Principal investigator:
Dr Margriet van Gestel
Lead institution:
Avans University of Applied Sciences, Netherlands

Our research project aims to develop a method for detecting Alzheimer’s disease early, using simple eye scans. Alzheimer’s is a devastating condition affecting memory and thinking, but diagnosing it can be difficult and expensive. By analyzing pictures of the back of the eye, we believe we can identify changes linked to Alzheimer’s, even before symptoms appear.

We’re using artificial intelligence, which allows computers to learn and recognize patterns. This AI will analyze thousands of eye scans to find common features associated with Alzheimer’s. By training the AI with this data, we hope to teach it to spot these features accurately in new eye scans.

This exploratory phase of the project will last for three years, during which we’ll develop and refine the AI model, test its accuracy, and explore how it could be used in real-world healthcare settings. We’ll work closely with experts in medicine, data science, and ethics to ensure our approach is safe, reliable, and ethical.

The potential impact of this research on public health is immense. Early detection of Alzheimer’s disease is crucial because it allows for earlier intervention and better
management of the condition. With current diagnostic methods being costly and invasive, our non-invasive approach using eye scans could revolutionize how
Alzheimer’s is detected, making it more accessible and affordable for everyone.

By identifying Alzheimer’s at an earlier stage, patients could receive treatment sooner, potentially slowing down the progression of the disease and improving their quality of life. Additionally, our method could reduce the burden on healthcare
systems by streamlining the diagnostic process and reducing costs associated with advanced testing.

Overall, our research has the potential to make a significant positive impact on public health by offering a simpler, more effective, and more accessible way to detect
Alzheimer’s disease early, ultimately leading to better outcomes for patients and their families.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/ai-model-for-the-early-detection-of-neurodegenerative-diseases-using-ocular-imagesfundus-oct-octa-etc-ct-mri-demographics-clinical-data

AI-model for the early detection of Neurodegenerative Diseases using Ocular Images(fundus, OCT, OCTA, etc ), CT, MRI, demographics, Clinical data.

Last updated:
ID:
706446
Start date:
30 April 2025
Project status:
Current
Principal investigator:
Professor Tanvir Alam
Lead institution:
Hamad Bin Khalifa University, Qatar

Research Question: We want to answer the question on how we can improve the early detection of Neurodegenerative Diseases using ocular image and other imaging modality. Many studies have been conducted on brain MRI. But very few studies have been done along this line using ocular images. We believe inclusion on ocular images (along with MRI, demographic and clinical information) will improve the early detection of neurodegenerative disease.

Scientific Rationale

A recent study, published in 2024, conducted based on retinal fundus image from UK Biobank, for the prediction of prevalence and incidence of Parkinson’s disease [2]. But the proposed model achieved around 70% accuracy. This indicates the fundus images can be used for the detection of neurogenerative diseases. Our hypothesis is that if we can incorporate such ocular imaging modalities as part of current clinical practice of neurodegenerative disease diagnosis, it may help to early diagnose a lot of patients

Objective:

The primary objective of this project is to develop, AI-solution for the early detection of neurogenerative disorder(ND) i.e., Parkinson’s disease (PD), Dementia (all subtypes of Dementia), Multiple sclerosis (MS), Age related Macular Degeneration (AMD), based on
(a) ocular image, i.e., fundus image, optical coherence tomography (OCT), optical coherence tomography angiography (OCTA)
(b) Other imaging modality CT, MRI, images
(c) Clinical data (collected at UKBiobank form blood, urine, saliva, etc).
(d) Demographics information

For this purpose we will case-control based study between case( Parkinson’s disease (PD), Dementia (all subtypes of Dementia, Vascular, Lewy Body, Frontotemporal, Mixed Dementia, ), Multiple sclerosis (MS)) vs. control (normal) , AMD setup.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/aid-pais-ai-driven-integration-of-multimodal-omics-and-clinical-data-for-enhanced-understanding-of-post-acute-infection-syndromes

AID-PAIS – AI Driven integration of multimodal Omics and Clinical data for Enhanced Understanding of Post-Acute Infection Syndromes

Last updated:
ID:
748709
Start date:
17 October 2025
Project status:
Current
Principal investigator:
Dr Dominik Wolff
Lead institution:
Hannover Medical School, Germany

Scientific rationale:
Current research highlights the need to explore the long-term consequences of infectious diseases like COVID-19, which in many cases are chronic symptoms such as fatigue, cognitive impairment, and other long-lasting health problems. These post-acute infection syndromes (PAIS) are scientifically recognized, but the pathomechanisms are not understood. To develop effective treatments and improve the quality of life of those affected, it is crucial to determine which subtypes of PAIS are caused by which pathogens and to identify involved pathways and biomarkers corresponding to these subtypes.
Objectives:
We aim to comprehensively investigate and understand the underlying molecular mechanisms of PAIS by applying state-of-the-art machine learning and statistical models on clinical, imaging and omics data. We focus on the development of advanced methods and software tools with multimodal integration on large-scale high-dimensional data as necessitated by the complexity of PAIS. Specialized machine learning and omics analysis techniques, including deep learning-based feature and base models, semi-supervised and positive-unlabeled learning, GWAS and QTL mapping, aim to identify unrecorded cases, subtypes, and corresponding genetic markers of PAIS. The obtained insights help to improve diagnostic accuracy, develop new therapeutic approaches, and promote a deeper understanding of disease mechanisms. Specific biomarkers help to better characterize PAIS and determine patient-specific subtypes in the future, significantly improving diagnosis and treatment of PAIS in the medium to long term.
Research questions:
How can the identification of unrecorded PAIS cases be improved through machine and deep learning models?
How can subtypes involving various pathogens and pathomechanisms be identified using the different methods?
What are the characteristics of the identified PAIS subtypes, their corresponding biomarkers and potential therapeutic targets?


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/air-pollution-and-dementia-exploring-genetic-cardiovascular-and-epigenetic-moderators-and-mechanisms

Air pollution and Dementia: Exploring genetic, cardiovascular, and epigenetic moderators and mechanisms

Last updated:
ID:
87096
Start date:
16 February 2023
Project status:
Current
Principal investigator:
Mr Otto-Emil Ilmari Jutila
Lead institution:
University of Edinburgh, Great Britain

Dementia is a growing public health crisis. The genetic makeup of the individual can increase dementia risk and the environment can increase the risk of dementia. Air pollution is increasingly understood to have an impact on brain health. Prior studies investigating the relationship between air pollution and dementia often do not include the impact of an individual’s genetic makeup on the relationship. The individual’s genetic risk to dementia could modify the effect of air pollution on dementia risk; genetic make-up may increase resilience or vulnerability to air pollution exposure.
Air pollution has been firmly established to be associated with cardiovascular disease (CVD) which in turn can impact dementia, therefore CVD or related risk factors may mediate the association between air pollution and dementia.
Furthermore, air pollution may accelerate biological aging in the brain via telomere shortening and alteration of DNA methylation.
The overall aim of this project is to investigate the relationship between air pollution and different types of the risk of dementia and on cognitive decline. To determine if CVD mediates the relationship between air pollution and dementia. To investigate the impact of genetics on the risk of air pollution on dementia. Furthermore, to determine the underlying disease mechanism between air pollution and dementia such as epigenetic changes and telomere length.

The duration of the project is part of my Ph.D. and will last 36 months.

A better understanding of the effect of air pollution on dementia risk could allow for improved public health interventions to lower the risk of dementia.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/air-pollution-and-incidence-of-breast-cancer-a-prospective-study-in-the-uk-biobank

Air pollution and incidence of breast cancer: a prospective study in the UK biobank.

Last updated:
ID:
141517
Start date:
8 November 2023
Project status:
Current
Principal investigator:
Professor Qiang Ding
Lead institution:
Nanjing Medical University, China

Breast cancer is the leading cause of cancer death in females and the incidence is increasing. The incidence and progression of breast cancer are influenced by a combination of genetic and environmental factors, including lifestyle and especially, air pollutants. Ambient air pollution (AAP) is a major risk factor for many diseases, however, evidence is sparse and inconclusive on the association between air pollution and breast cancer. Solid and thorough population researches of this issue are needed. We aimed to assess the association of AAP including (PM2.5, PM10, NO2, SO2, CO and O3) with breast cancer risk and compared the breast cancer risk attributable to AAP and other established risk factors. We plan to conduct these comprehensive analyses by running this project for 3 year (2024-2027). Findings regarding environment factors have a significant public health implication in the prevention of breast cancer, reduce the incidence and mortality of breast cancer, and reduce the medical burden of social funds.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/air-pollution-greenness-and-their-interplay-with-dna-repair-genes-in-relation-to-breast-cancer

Air pollution, greenness and their interplay with DNA repair genes in relation to breast cancer

Last updated:
ID:
67356
Start date:
25 November 2020
Project status:
Current
Principal investigator:
Mrs Carmen Smotherman
Lead institution:
University of Florida, United States of America

In this study, we aim to explore associations in adult women between exposure to air pollution and greenness and breast cancer risk. In addition, we will explore if the variation in genes responsible for the DNA repair affect these associations. Breast cancer remains the most common cancer among women worldwide and in the United States. Known risk factors for breast cancer, however, explain only about 50% of variation in breast cancer risk. Among potential environmental risk factors for breast cancer, air pollution has been getting attention recently as possible contributor to breast cancer, though the overall evidence remains inconsistent. On the other hand, exposure to green environments can reduce adverse environmental exposures such as air pollution, noise, and extreme heat, as well as promote physical activity and reduce stress, all of which could influence health outcomes including cancer. However, only three studies examined associations of greenness with breast cancer. Finally, none of the previous studies on air pollution and BCa included variations in the genes responsible for DNA damage repair. We anticipate completing the project in up to 3 years after the approval. Due to a large proportion of women living in the areas with unfavorable air quality, our results could generate new approaches for incorporating air pollution into breast cancer risk assessment or tailored active surveillance or prevention strategies in women with high exposure level.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/air-pollution-immunohematologic-failure-and-cardiovascular-risk

Air pollution, immunohematologic failure, and cardiovascular risk

Last updated:
ID:
61548
Start date:
12 January 2021
Project status:
Current
Principal investigator:
Dr Sadeer G Al-Kindi
Lead institution:
University Hospitals Cleveland Medical Center, United States of America

Aim: The aim of this proposal is to understand the impact of non-traditional factors (immune dysfunction, air pollution, and socioeconomic status) on risk of heart disease.
Scientific Rationale: Air pollution and socioeconomic status have been individually associated with risk of heart disease. We have shown that dysfunction of the immune system (reflected by reduced number of lymphocytes) and the red blood cell system (as measured by high variability in red cell size) are associated with risk of death from heart disease
Project Duration: The duration of this project is approximately 3 years
Public Health Impact: Understanding non-traditional risk factors of heart disease will help identify high-risk populations, for whom intensive cardiovascular risk reduction can be investigated.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/air-pollution-lifestyle-genetic-susceptibility-in-relation-to-the-risk-and-mortality-of-urologic-cancers

Air pollution, lifestyle, genetic susceptibility, in relation to the risk and mortality of urologic cancers

Last updated:
ID:
94258
Start date:
15 April 2024
Project status:
Current
Principal investigator:
Dr Benjamin Chung
Lead institution:
Stanford University, United States of America

Along with the aging population and socioeconomic development of the past few decades, the incidence and mortality rates of urologic cancer are increasing at an alarming rate around the world. Urologic cancers, which mainly include prostate, bladder, and kidney cancer, contribute to ~13% of total new cancer diagnoses globally. The differences in urologic cancer outcomes between industrialized and developing nations continue to grow. As one of the most significant urologic diseases, kidney cancer is predicted to cost $1.6 billion (2006USD) and cause over 131,000 deaths worldwide. However, the etiology of urologic malignancies and the complicated impact of environmental exposures remains poorly understood.

Previous studies have suggested relationships between air pollution or dietary factors and certain urologic cancers. Currently, only a few of these factors, such as a specific pollutant or dietary behavior, have been found to be associated with the risk of certain urologic cancers, and the corresponding interventions have been adopted individually. However, the results are inconsistent, and their paired and complex interactions are largely unknown. Consequently, we intend to utilize the urologic cancer-related data, air pollution data, and lifestyle data from the UK biobank to investigate the major risk factors. The specific objectives are as follows:1) investigate the association of (a) individual air pollutants and the complex air pollution mixtures; (b) diet and nutrition and the mixtures of those consumptions; (c) biochemistry markers, telomere length, and dual-energy X-ray absorptiometry (DXA) scan body composition measurement, with the incidence and mortality of urologic cancers, including prostate, kidney, and bladder cancers; 2) evaluate the interactions between air pollutants and diet/nutrition in relation to the incidence and mortality of urologic cancers. We also would like to extend our scope to explore Aim 1a/1b stratified by status of genetic susceptibility, lifestyle factors, and specific health status (e.g., hypertension and diabetes) and medication usage, as well as to compare the hospitalization admission and primary health care utilization rates in patients with different tumor stages of urologic cancers before, during, and after COVID-19 lockdown.

The findings will eventually lead to the enhancement of screening, prevention, and early intervention in urologic cancer patients. We expect to finish this study and send publications to international peer-review journals for review within 36 months of receiving the data, at which point we will initiate analyses. We expect this study to offer a profile of environmental, dietary, and mixture exposures and urologic cancer risk.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/air-pollution-noise-and-cardio-respiratory-diseases-bioshare-environmental-core-project

Air Pollution, Noise and Cardio-respiratory diseases-BioSHaRE Environmental Core Project

Last updated:
ID:
5179
Start date:
6 January 2014
Project status:
Closed
Principal investigator:
Yutong Cai
Lead institution:
Imperial College London, Great Britain

Research has reported harmful effects of ambient air pollution and road traffic noise on the human cardiorespiratory system. However, there are few studies to date looking at these two environmental risk factors jointly to investigate the cardiovascular and respiratory effects. This proposed project aims to quantify the separate and joint effects of air pollution and noise exposure on cardiovascular and respiratory outcomes at the individual level, specifically cardiovascular diseases (ICD-10 codes I00-I99), Asthma (ICD-10 codes J45) and levels of bio-chemical markers for cardiovascular diseases including blood lipids, C-reactive protein, blood glucose.
We propose to use the whole cohort of UK Biobank to address our research questions. Questionnaire data including demographic, socioeconomic, and lifestyle data will be requested. Health outcome data will be ascertained both on the basis of self-report and medical records. Inpatient data with regards to all incident hospital admissions according to primary and underlying admissions causes coded under ICD-10 I00-I99 (cardiovascular diseases) will be requested. We also request data from death registry linkage-coded according to underlying (primary) and contributory (secondary) ICD-10 I00-I99. The asthma outcome will be ascertained by self-reported data at this stage. These data will be harmonised in line with those from four other cohorts involved in this BioSHaRE-funded project, namely HUNT(Norway), Lifelines (the Netherlands), EPIC-Oxford (UK), EPIC-Turin (Italy). Environmental exposure data (air pollution and noise) at the time of recruitment were created through linkage of individual address data to geocoded databases at Imperial College,London. The ultimate objective is to pool harmonised data from all these five Biobanks including UK Biobank for epidemiological analysis.
This project fits well with the UK Biobank?s central aim. It will help better understand the environmental determinants of cardio-respiratory disease by disentangling the health effects of these two exposures and thereby inform targeted preventive strategies and contribute to scientific knowledge.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/airborne-adversaries-investigating-the-impact-of-air-pollution-on-sarcopenia-development-and-progression

Airborne Adversaries: Investigating the Impact of Air Pollution on Sarcopenia Development and Progression

Last updated:
ID:
303535
Start date:
21 February 2025
Project status:
Current
Principal investigator:
Professor Chaohui Yin
Lead institution:
Henan Agricultural University, China

This research project seeks to explore the impact of air pollution on sarcopenia, a condition characterized by the progressive loss of skeletal muscle mass and strength, predominantly affecting older adults. The primary aim is to determine how exposure to air pollutants, such as particulate matter and chemical toxins, contributes to the development and acceleration of sarcopenia.
Scientific Rationale:
Current evidence suggests that air pollution can have widespread effects on human health, including respiratory and cardiovascular diseases. However, its influence on muscle health is less understood. Muscles are critical for maintaining mobility, stability, and overall energy in older adults. Therefore, understanding how environmental factors like air pollution affect muscle tissue is crucial for developing strategies to protect and enhance muscular health as people age.
Project Duration:
The study is designed to span three years. The first year will focus on collecting and analyzing existing data on air pollution levels and health outcomes related to muscle mass and strength. In the second year, we will conduct observational studies and possibly recruit participants for trials to assess direct biomarkers of muscle health in polluted environments. The final year will involve synthesizing data, forming conclusions, and publishing the findings.
Public Health Impact:
The implications of this research are significant for public health. By establishing a clear link between air pollution and muscle health, this project can lead to better health advisories and more robust public health policies aimed at reducing exposure to harmful pollutants, particularly in urban areas. Furthermore, the research could foster the development of community health programs that focus on preventive measures to combat sarcopenia, such as dietary recommendations, exercise regimes, and environmental health awareness campaigns. These initiatives could particularly benefit aging populations, helping them maintain healthier, more active lifestyles despite environmental risks. Ultimately, the research aims not only to enhance scientific understanding but also to foster real-world applications that improve quality of life and health outcomes for older adults.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/alcohol-consumption-and-brain-health

Alcohol consumption and brain health

Last updated:
ID:
55929
Start date:
9 March 2020
Project status:
Current
Principal investigator:
Dr Anya Topiwala
Lead institution:
University of Oxford, Great Britain

Alcohol consumption in endemic in our society, with almost 40% of the world’s population drinking. Whilst we know that very heavy drinking is detrimental to the brain, and can lead to dementia, more moderate intakes have been claimed to be protective. However, there were limitations in the way this research was carried out and the results may be spurious. Given the societal burden of dementia, identification of any modifiable factor such as alcohol consumption, would have grave public health implications.

UK Biobank contains information on the drinking habits of hundreds of thousands of people in the UK, and is the largest brain imaging resource worldwide. This offers for the first time the opportunity to investigate if and how alcohol impacts brain structure and function.

The specific aims are:
1. Does alcohol cause dementia?
2. Does drinking damage brain structure?
3. What are the pathways to damage?

Better understanding of these issues will hopefully identify ways to intervene, with a potentially huge public health impact.
The project duration is 5 years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/alcohol-consumption-and-chronic-health-conditions-examined-through-non-linear-mendelian-randomisation-analysis

Alcohol consumption and chronic health conditions examined through non-linear Mendelian Randomisation analysis

Last updated:
ID:
86004
Start date:
30 May 2022
Project status:
Closed
Principal investigator:
Ms Rachel Visontay
Lead institution:
University of Sydney, Australia

Heavy alcohol consumption is known to increase the risk for a wide range of diseases, leading to significant financial and societal costs. However, research also tends to find that moderate consumption is actually protective against certain health outcomes when compared to abstinence. Despite increased work in recent years, it is still unclear whether this association is a true causal relationship or simply a by-product of methodological limitations. Mendelian Randomisation – a method that studies genetic tendencies to drink in place of drinking itself – allows for determining whether a causal relationship exists, and thanks to more recent developments, what shape this relationship takes. This study aims to apply this method to the examination of various alcohol-long-term health relationships using the rich genetic and health data offered by the UK biobank. We will also conduct more conventional analyses of these relationships so that results can be compared. It is estimated that this work will take 36 months to complete. Given that evidence about alcohol’s protective effects is often incorporated into national safe drinking guidelines and public health policy, determining whether these effects are truly causal stands to have a significant impact.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/alcohol-consumption-and-the-risk-of-vascular-outcomes

Alcohol consumption and the risk of vascular outcomes

Last updated:
ID:
21886
Start date:
20 July 2016
Project status:
Closed
Principal investigator:
Angela Wood
Lead institution:
University of Cambridge, Great Britain

Aim: To determine risk of various vascular diseases and all-cause mortality with alcohol consumption

There is debate about whether moderate alcohol consumption reduces or increases risk of cardiovascular disease. To improve the validity of comparisons, we will assess the shape, specificity, magnitude and independence of associations between alcohol consumption and risk of various vascular outcomes and all cause mortality.
This proposed research will yield reliable findings with potentially important implications for public health with regards to recommended drinking levels for the prevention of vascular disease.

Data from UK Biobank will be combined with individual-participant data on 300,238 current drinkers from 91 prospective studies (18,326 cardiovascular events) from the Emerging Risk Factors Collaboration and with 24,000 current drinkers from EPIC-CVD.

We will perform a meta-analysis of individual-participant data from multiple studies to calculate hazard ratios across pre-defined categories of long-term average alcohol consumption for different fatal cardiovascular diseases and for non-fatal myocardial infarction and cerebrovascular disease. The analyses will be restricted to those without a history of cardiovascular disease and current drinkers. We estimate this to be approximately 261,000 participants.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/alcohol-non-communicable-disease-mendelian-randomisation-consortium

Alcohol Non-communicable Disease Mendelian Randomisation Consortium

Last updated:
ID:
24619
Start date:
1 August 2016
Project status:
Closed
Principal investigator:
Dr Caroline Dale
Lead institution:
University College London, Great Britain

We aim to reveal the true nature of the association of alcohol with non-communicable diseases, including cardiovascular disease and dementia/ cognitive impairment, by using genes as instrumental variables. Alcohol is the third leading risk factor for death and disability worldwide. While the harmful effects of alcohol on many health conditions are well-established, uncertainty remains concerning the reported protective effects of light-to-moderate alcohol consumption for cardiovascular disease. This research seeks to clarify the true causal nature of the association and inform public health policy for this important global risk factor. Existing evidence for the cardioprotective effect of light drinking comes from people reporting alcohol consumption and subsequent follow-up of events many years later. However, it is well know that people inaccurately report alcohol consumption and some change their drinking behaviour after being diagnosed with disease. In addition, people who drink small amounts of alcohol also tend to have other healthy behaviours (confounding). We plan to use a different strategy previously described as similar to a Randomised Control Trial using a genetic variant that tends to make people drink less to explore associations with disease (Mendelian Randomization). The full Biobank cohort will be included to maximise statistical power for genetic analyses.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/algorithmic-fairness-and-bias-in-the-prediction-of-cardiometabolic-and-brain-health-related-outcomes

Algorithmic fairness and bias in the prediction of cardiometabolic and brain health-related outcomes

Last updated:
ID:
103919
Start date:
17 October 2023
Project status:
Current
Principal investigator:
Dr Tibor V. Varga
Lead institution:
University of Copenhagen, Denmark

This research project aims to answer important questions about the fairness of current risk scores used to predict cardiometabolic (type 2 diabetes, heart disease) and brain-related diseases (neurological disorders, depression, anxiety). We want to find out if these models, which were largely developed in majority populations, work equally well for e.g., minority groups with higher disease burdens or for population subgroups who are otherwise disadvantaged. Additionally, we want to explore the possibility of creating new prediction models that are not only accurate but also fair.

In the first part of the project, we will analyze existing prediction models to see if they are unfair when it comes to predicting diseases. This will help us understand if certain groups are being treated differently or receiving sub-standard care. In the second part, we will develop new prediction models that take into account a wide range of factors, such as demographics, socioeconomic status, diet, lifestyle, and sleep patterns, and other health-related information, such as medication use and disease histories. We will train these new models to perform equally well for different population groups defined by race/ethnicity, sex, and socioeconomic factors like income and education.

The rationale for this research is based on the fact that people with lower socioeconomic status and certain cardiometabolic diseases often face healthcare inequalities, experience more severe complications, and have shorter lifespans. These inequalities not only have a negative impact on individuals but also carry significant costs for governments. By improving the fairness of healthcare processes, we can improve lives and benefit economies.

Currently, many prediction models are developed based on data from majority populations, which may not work as well for minority or marginalized groups. Moreover, if these models are trained on biased data, they can perpetuate existing biases and inequalities.

By developing a comprehensive framework to assess algorithmic fairness and identifying biases in existing models, we can pave the way for new fair models that will help address healthcare gaps. The resulting predictive models will consider performance, fairness, and explainability, and have the potential to replace current risk assessment algorithms and improve preventive strategies for cardiometabolic diseases.

This research project is expected to last for three years and has the potential to make a significant impact on public health by promoting fair and accurate prediction models for cardiometabolic outcomes.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/altered-connectivity-between-the-two-brain-hemispheres-in-the-depressive-personality

Altered connectivity between the two brain hemispheres in the depressive personality

Last updated:
ID:
48507
Start date:
24 December 2019
Project status:
Closed
Principal investigator:
Dr Xuesong Li
Lead institution:
Beijing Institute of Technology, China

Altered connectivity between the two brain hemispheres has been found in mental disorders. Previous study of our group has shown decreased between-hemispheres connectivity in patients with late-life depression relative healthy controls. However, systematic study is still lacking regarding which factors are related to the between-hemisphere connectivity reduction: cerebrovascular disorders, clinical symptoms, gender, personality traits, genetic factors or some key life-style factors. In this study, we propose to combine brain structural and functional MRI data with other non-imaging data such as depression-related genetic variants, personality traits, mood and anxiety as well as other clinical symptoms, cognitive function, life-styles, gender, blood pressure, and gait speed etc to examine which of these factors is highly associated with reduced between-hemisphere connectivity in major depression. Given the vital role of corpus callosum in connecting the two hemispheres, we will further subdivide the corpus callosum into small circles with 5mm in diameter and 5 mm apart between any two sub-regions. Between-hemisphere connectivity is a key neural connectivity feature vital for many behaviors, yet has been relatively neglected. Our project will not only identify factors related to the reduction of between-hemisphere connectivity, but also can determine the related gene loci regulating the connection between the hemispheres in healthy controls but genetic variants that contribute to the reduction of between-hemisphere connectivity and depression. The moderation/modulation effects among these factors will also be examined.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/alzheimers-disease-and-immune-events

Alzheimer?s Disease and Immune Events

Last updated:
ID:
15181
Start date:
1 October 2015
Project status:
Current
Principal investigator:
Dr Laura Marie Winchester
Lead institution:
University of Oxford, Great Britain

Our aim is understanding the role of the immune system in cognition and Alzheimer?s disease (AD). To reach this goal, we want to analyse how a number of exposures (genes and events of the immune system) affect AD outcome (and cognitive function as a proxy), and then identify the mechanisms of these effects (e.g. brain morphology, circulatory system). Some of our exposures (e.g. CLU, CD33, DKK1, NSAIDs) have a proven (but not understood) link with AD, other exposures are related events (e.g. cancer, oral infection). Our target mechanisms are all potential stages where this link materialises. We think understanding the role of the immune system in AD is the essential step towards understanding AD. The best approach for this is to systematically investigate how immune genes and events affect AD and cognition, and UK-Biobank contains the essential variables required for such a systematic investigation (i.e. simultaneously recorded genes and medical history). Our investigation will render results closely matching UK-Biobank purposes: (1) understanding the role of the immune system in AD; (2) finding novel early immune biomarkers of AD; (3) developing statistical tools to better analyse large and multimodal medical datasets. As an informatics team, we have a range of skill sets and we plan to divide the research accordingly. Participant clinical measurements will be mined by Dr Chi-Hun Kim with a particular focus on linking immunologic events with cognition and multimodal outcomes. Genomic data will then be compared to relevant characteristics at a genome-wide level and focusing on immune gene sets of interest by Dr Laura Winchester. Overseeing and machine learning analysis will be implemented by Dr Alejo Nevado-Holgado. We intend to implement a full cohort analysis. From the full cohort, the effects of immune genes and events will be tested on cognitive tests and the diagnosis and onset of AD. We will approach this question from four fronts: GWAS (all SNPs needed), PheWAS (most phenotypes needed), pathway analysis and factor analysis (to investigate the causal factors in the multilevel sense). For this reason, we think it is vital to have access to the full UK-Biobank cohort with set variables.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/alzheimers-disease-genetics-consortium-multi-ethnic-alzheimer-disease-genetics-analysis

Alzheimer’s Disease Genetics Consortium Multi-ethnic Alzheimer disease Genetics Analysis

Last updated:
ID:
64211
Start date:
11 January 2021
Project status:
Current
Principal investigator:
Dr Farid Rajabli
Lead institution:
University of Miami, United States of America

Our goal in this study is to evaluate the role of race/ethnicity and ancestry-specific genetic factors in known Alzheimer disease (AD) genes in our multi-ethnic dataset and to identify novel risk loci correlated with genetic ancestry. In the experimental design, we will perform a series of ancestry-aware statistical approaches to characterize the influence of ancestral background located in or near the genomic region of known genes in AD. Furthermore, to identify novel genes specifically through under-studied ancestries, we will perform genome-wide analysis by testing the genetic factors in the context of genetic ancestry. This study will systematically assess the role of genetic ancestry on AD risk in a diverse set of multi-ethnic samples, potentially leading to a set of genes and genomic regions that modify existing AD risk factors. This gives us the opportunity to identify novel factors influencing AD that may contribute to health disparities. This study will provide insights into the risk factors correlated with genetic ancestry and will enhance the power and extend studies in under-studied populations. In particular, identification of population-specific variations that influence disease could inform precision medicine initiatives, and lead to the development of ancestry-specific AD treatments. This would improve treatments, and help reduce health disparities.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/alzheimers-disease-model-a-hybrid-quantitative-systems-pharmacology-qsp-ai-driven-evidence-platform-for-rd

Alzheimer’s Disease Model: A Hybrid Quantitative Systems Pharmacology (QSP) & AI-Driven Evidence Platform for R&D

Last updated:
ID:
58339
Start date:
17 June 2020
Project status:
Closed
Principal investigator:
Dr Alexander Knight
Lead institution:
Holmusk Europe Limited, Great Britain

Alzheimer’s disease (AD) is the most common form of dementia, affecting patients’ ability to remember and to perform everyday tasks. One million people in the UK will have dementia by 2025, and this will increase to two million by 2050. 60% of these dementia cases are caused by Alzheimer’s disease. The current cost of dementia in the UK is £26bn and this is predicted to rise to £55bn by 2040. Globally, there are 50 million dementia sufferers, and again, this is predicted to rise dramatically, to 152m by 2050.

AD has proved hard to treat; the last 57 trials of new drugs have been unsuccessful. One of the reasons for this seems to be that scientists do not fully understand the way that the disease progresses in the brain. Many different types of data have been collected, including brain scans and blood tests, from many patients and from healthy people. This data probably contains many useful clues to understanding the disease, and how best to treat it. But the data sets are big, complex and hard for humans to interpret.

New methods in “machine learning” and “artificial intelligence” are a potential solution. They enable a computer to look for patterns in the data that a human might miss. We have previously used these tools to build a model of how AD progresses in the brain and affects the brain’s function. The aim of the project we are proposing is to use the data from the UK Biobank to improve our model so that we can make more accurate predictions and improve our understanding of how the disease can be treated. We anticipate that this work will take approximately 2 years because of the complexity of the data sets. We will then make our model available to companies developing new drugs and treatments for AD. In the long run, the hope is that new treatments will be available to slow or stop the progression of the disease, and possibly even to prevent it.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/ambient-temperature-exposure-and-systemic-inflammation-large-scale-proteomic-insights-from-uk-biobank-olink-data

Ambient temperature exposure and systemic inflammation: large-scale proteomic insights from UK Biobank Olink data

Last updated:
ID:
1008735
Start date:
2 October 2025
Project status:
Current
Principal investigator:
Mrs Bo Sun
Lead institution:
IUF-Leibniz Research Institute for Environmental Medicine, Germany

Research outline: This project will examine how fluctuations in ambient temperature influence systemic inflammation using UK Biobank data. We aim to replicate evidence from smaller studies showing that cold and hot weather alter inflammatory biomarkers1,2, and extend these findings in a much larger, more diverse population. The unique UK Biobank Olink dataset, which includes the Target 96 and Explore Inflammation panels (~700 unique proteins), provides high-quality proteomic measurements with extensive participant and environmental data, offering a unique opportunity to identify which inflammation-related proteins are most sensitive to temperature changes.

Research questions and aims: We will investigate (1) which specific inflammatory proteins are most strongly associated with recent temperature variation; (2) the shape and strength of these relationships over short-term (days) and medium-term (weeks to months) windows; and (3) whether associations differ by individual characteristics (e.g., age, sex, health) or environmental context (high vs. low air pollution).

Scientific rationale: Extreme heat and cold increase health risks, but the biological pathways remain unclear. Temperature stress may trigger systemic inflammation. Prior studies support this: colder weather was linked to higher levels of inflammatory proteins1, and some biomarkers responded to heat 2, but these studies were limited in size and scope. Leveraging the scale and diversity of UK Biobank Olink data, we can detect subtle temperature-biomarker associations, validate previous findings, and identify vulnerable subgroups. These insights will advance understanding of climate-related health impacts and links to outcomes such as cardiovascular events, kidney dysfunction, and other inflammation-driven conditions, informing strategies to protect at-risk populations.
1. Ni, W., et al. Environ Sci Technol. DOI:10.1021/acs.est.3c00302
2. Sun, B., et al. Environ Res. DOI:10.1016/j.envres.2025.122382


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/amplitude-of-low-frequency-fluctuation-in-resting-state-mri-signal-in-hypertensives

Amplitude of low frequency fluctuation in resting state MRI signal in hypertensives

Last updated:
ID:
73771
Start date:
9 August 2021
Project status:
Current
Principal investigator:
Dr Owen Woodward
Lead institution:
Cardiff University, Great Britain

High blood pressure is a major risk factor for several diseases, including stroke, heart disease and dementia. In the majority of cases, it is not known what causes high blood pressure. One proposed cause is an increase in the fight-or-flight response of the body. This increase would serve to drive blood pressure up in an effort to maintain blood flow to the brain. The fight-or-flight response is regulated by various parts of the brain. Magnetic resonance imaging (MRI) uses strong magnetic fields to take images of the brain. Functional MRI (fMRI) can be used to investigate changes in neural activity and blood flow to the brain. In this study, we propose to use low frequency fluctuations in the fMRI signal as a marker of vascular function. We will use this to compare the function of the fight-or-flight response between people with and without high blood pressure. This will hopefully lead to a better understanding of the cause of high blood pressure, and ultimately to better treatment of the disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/amyloid-and-tau-status-prediction-with-clinical-and-cognitive-data-an-explainable-machine-learning-approach

Amyloid and tau status prediction with clinical and cognitive data: An explainable machine learning approach.

Last updated:
ID:
358299
Start date:
28 October 2024
Project status:
Current
Principal investigator:
Professor Wyllians Vendramini Borelli
Lead institution:
Federal University of Rio Grande do Sul, Brazil

This study aims to investigate the probability of presenting Alzheimer’s disease without performing any diagnostic test. Ideally, we will be able to predict Alzheimer’s disease pathology after a single medical consultation – in other words, this study will provide a tool to measure the probability of presenting Alzheimer’s disease pathology using only information that you provide to your doctor (such as your age, sex, family history of dementia). We will conduct a study using a machine learning method that can be adapted to the context used. The model can be optimized for public health policies in identifying individuals with a high probability of presenting Alzheimer’s disease – and targeting this to treat and prevent worsening.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-adaptive-artificial-intelligence-approach-to-improve-efficiency-and-quality-in-cardiac-mri

An Adaptive Artificial Intelligence Approach to Improve Efficiency and Quality in Cardiac MRI

Last updated:
ID:
89477
Start date:
15 September 2022
Project status:
Current
Principal investigator:
Dr Kavitha Vimalesvaran
Lead institution:
Imperial College London, Great Britain

Aim:
This project aims to reduce the numbers of visits that patients and doctors need to spend waiting for and taking images of the heart. It aims to this by developing AI-based tools for use during the scanning of patients in order to ensure that all necessary data is acquired in order to make more confident diagnoses about potential heart problems within a shorter timeframe.

Scientific rationale:
The recent use of AI in healthcare has been dominated by demonstrations of performance in analysing highly curated imaging data which has, to date, only rarely been translated into clinically deployable technology. This project considers the comparatively large burden on healthcare systems (both financially and in terms of human capital) and individual patients (in terms of anxiety and inconvenience) of obtaining the images optimally.

Project duration:
24 months

Public health impact:
The expected value us a reduction in scan times to allow more patients to benefit from cardiac magnetic resonance imaging, improved confidence in diagnosis (to improve care) and improved patient experience (by avoiding prolonged or unnecessary scans). Over the next decade, it is intended that it will be utilised alongside real-world practice and have a tangible effect on healthcare productivity and patient care.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-ai-based-decision-support-system-for-early-diagnosis-of-hematologic-malignancies

An AI-based decision support system for early diagnosis of hematologic malignancies

Last updated:
ID:
171989
Start date:
4 December 2024
Project status:
Current
Principal investigator:
Dr Adel Elomri
Lead institution:
Hamad Bin Khalifa University, Qatar

Blood cancer is one of the deadliest types of malignancies. If not diagnosed and treated early, its disease progression can lead to death in both adults and children. In fact, it was proven that most of the cases are treatable if detected early. Therefore, a need for an accurate diagnostic system to early spot the signs of blood cancer is crucial. The expected duration of this PhD student project is 1 year. This study’s novelty stems from its exclusive use of CBC, paving the way for a cost-effective digital decision support system (DSS) for blood cancer diagnosis. This approach facilitates broad population screenings, optimizes referrals in primary and secondary care settings, and promises timely diagnoses in specialized centers.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-artificial-intelligence-algorithm-based-on-deep-learning-and-molecular-diagnosis-for-identifying-ocular-diseases

An artificial intelligence algorithm based on deep learning and molecular diagnosis for identifying ocular diseases.

Last updated:
ID:
45270
Start date:
18 December 2019
Project status:
Current
Principal investigator:
Professor Jianzhong Su
Lead institution:
Eye Hospital of Wenzhou Medical University, China

We plan to construct a clinically useful model to identify several ocular diseases automatically, and improve the efficiency of diagnosis.
Artificial intelligence (AI) has the potential to revolutionize disease diagnosis and management by performing classification difficult for human experts. Within ophthalmology, artificial intelligence is already augmenting diagnostic imaging capabilities, which may soon lead to deployment of cost-efficient telemedicine screening programs worldwide. The majority of these early efforts have focused on the analysis of color fundus photographs or optical coherence tomography (OCT) scans for detection of posterior segment diseases such as diabetic retinopathy, age-related macular degeneration, and glaucoma. Deep learning refers to a subset of artificial intelligence, composed of algorithms that use a cascade of multilayered artificial neural networks for feature extraction and transformation. Drawing inspiration from the structure of the human mind, convolutional neural networks consist of thousands of individual neurons capable of performing complex tasks, such as image recognition and classification, based on pixel or voxel intensity. Each successive layer in the network uses the output from the previous layer as input, with the final layer revealing the diagnostic output. There are a lot of studies for retinal image classification selected binary classification. But the results of studies about multi-categorical retinal image classification were not as good as binary classification. In the other hand, many ocular diseases have been considered genetically defined complex disorders such as AMD and DR. For example, with 50% or more of the heritability of AMD already explained by two major loci harboring coding and non-coding variation at chromosomes 1q (CFH) and 10q (ARMS2/HTRA1). So it’s available to identify some ocular diseases through genetic classifier.
We expect to finish this project in 3 years. If all goes well, this study will increase efficiency of medical diagnosis, reduce barriers to access for areas where an eye care provider may not be present, provide earlier detection of referable eye disease, improve prognosis and decreasing healthcare costs through earlier intervention of treatable disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-artificial-intelligence-based-approach-for-the-early-detection-of-chronic-kidney-diseases-from-digital-retinal-photographs

An artificial intelligence-based approach for the early detection of chronic kidney diseases from digital retinal photographs

Last updated:
ID:
106155
Start date:
20 September 2023
Project status:
Current
Principal investigator:
Professor Ahsan Habib Khandoker
Lead institution:
Khalifa University of Science and Technology, United Arab Emirates

Chronic Kidney Disease (CKD) is a serious condition that can lead to kidney failure. Diabetes is the leading cause of CKD, and people with diabetes are advised to get regular eye exams to monitor for signs of retinopathy, which is linked to CKD. To make an early diagnosis of CKD easier, we can use artificial intelligence to analyze retinal images and predict the onset and stage of CKD. We will use deep-learning models to analyze the retinal images and collect clinical data from patients, such as age, blood pressure, and diabetes status, to predict their kidney function. We will use this information to train separate models to predict kidney function and blood glucose levels. Then, we will use these predicted values to detect CKD and its stages. Finally, we will combine the retinal image data and clinical information to create a more accurate model. Early detection is appealing because inexpensive intervention can ameliorate renal function early and prevent continuous deterioration. The shortcomings of traditional analysis methods make a new comprehensive assessment approach urgently needed in kidney disease patients and a large population scale. ML and DL combined with retinal imaging is a relatively prominent frontier in this research area.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-atlas-of-clinical-associations-mapping-to-variants-within-protein-structures

An Atlas of Clinical Associations Mapping to Variants within Protein Structures

Last updated:
ID:
61255
Start date:
16 June 2021
Project status:
Current
Principal investigator:
Dr Dana Colleen Crawford
Lead institution:
Case Western Reserve University, United States of America

Each human genome consists of approximately 6 billion pairs of DNA, represented by combinations of nucleotides known as A, T, C, and, G. Only a fraction of this genome represents genes, units of As, Ts, Cs, and Gs that instruct the cell to make something like a protein. We know from rare genetic diseases like sickle cell anemia that a single change in the DNA sequence can cause disease by altering the protein’s function. In general, the DNA sequences of genes that result in protein products are thought to be very important and, when altered, could be a cause of severe disease or death early in life or make a person more susceptible to developing a chronic disease later in life.
The genomics community now has the ability to sequence genomes, which gives us the complete catalog of a person’s As, Ts, Cs, and Gs. While we have a good idea of where genes are located in these sequences, we do not understand how a single change of an A, T, C, or G within the gene will impact the making of or function of the gene’s protein. We also do not have an understanding if these changes within the protein-important sequences will make people more susceptible to disease and, if so, which ones.
We expect the UK BioBank dataset will provide us with a list of DNA changes in protein-important sequences that possibly predispose people to specific common diseases. The UK BioBank is one of the only studies available to us that has DNA sequences in protein-important regions of the genome on people who have also provided health-related data and blood for laboratory measurements for important markers of health such as cholesterol levels. We have developed a new way to screen all the As, Ts, Cs, and Gs to identify which changes might be bad for the protein. Using our new method, we will screen the UK BioBank sequences for these changes and test whether or not these sequence changes are more frequent among UK BioBank participants with diseases compared to healthy UK BioBank participants. Results from this study will help the scientific community better understand which genetic changes are important for common human diseases and may provide us with clues as to how to better prevent, diagnose, and/or treat them.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-epidemiological-and-genetic-analysis-of-traits-associated-with-musculoskeletal-conditions

An epidemiological and genetic analysis of traits associated with musculoskeletal conditions

Last updated:
ID:
58855
Start date:
19 August 2020
Project status:
Current
Principal investigator:
Dr Francis S Collins
Lead institution:
National Human Genome Research Institute, United States of America

Musculoskeletal (MSK) conditions such as weakness, chronic pain, arthritis, osteoporosis, and fibromyalgia affect up to one in four people across the UK. MSK conditions are a leading contributor to disability worldwide and are commonly linked to an increased risk of developing other chronic health conditions. Due to a poor understanding of the underlying pathophysiology, current treatments are not very effective, creating a sense of urgency surrounding MSK research.

To better understand the causes of MSK conditions, we will conduct a study focused on MSK-relevant physiological and imaging measures in the UK Biobank. Using machine learning techniques, we intend to derive novel measures that are medically relevant to MSK conditions and conduct an epidemiological and genetic analysis of these MSK measures. We will integrate our results with previous studies in order to improve our understanding of the causes of MSK conditions. We estimate that this study will take up to three years. Collectively, our study has the potential to help improve our understanding of the cause of MSK conditions and contribute to efforts to prevent, diagnose, and treat MSK conditions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-epidemiological-and-genome-wide-association-approach-to-coeliac-disease

An epidemiological and genome-wide association approach to coeliac disease

Last updated:
ID:
18532
Start date:
1 August 2016
Project status:
Current
Principal investigator:
Professor Elizabeth Soilleux
Lead institution:
University of Oxford, Great Britain

Coeliac disease (CD) is an autoimmune condition of the digestive system characterised by an adverse reaction to gluten. CD prevalence is estimated at 1% in Europe and North America, but may be as high as 8-10%, whilst UK CD incidence has increased fourfold between 1990 and 2011, likely due to increased diagnostic testing. The current proposal has two main aims: firstly, to investigate the association between a wide range of sociodemographic, environmental and lifestyle in association with prevalent and incident CD; secondly, to perform a genome-wide association study (GWAS) in order to identify genetic variants associated with CD. CD consists of a range of debilitating gastrointestinal and non-gastrointestinal symptoms. Whilst avoidance of gluten can ameliorate these symptoms, evidence suggests that CD is underdiagnosed or often misdiagnosed as other conditions such as irritable bowel syndrome (IBS). Furthermore, apart from the presence of HLA-DQ2 or HLA-DQ8 genotypes, there is a lack of established factors that increase the risk of CD or exacerbate the condition. This research project into the lifestyle, sociodemographic and genetic determinants and distribution of CD will ultimately provide novel insights that will inform the prevention, treatment and diagnosis of the condition. We will investigate the epidemiology of CD through exploring how it varies by environmental and sociodemographic characteristics, such as smoking, alcohol, diet, early-life exposure etc., and biochemical markers. We will undertake a GWAS in order to identify variants associated with CD which will provide an insight into potential mechanisms. A key aspect of our research is establishing the characteristics that distinguish CD from conditions with similar symptomatology. Therefore, we will also perform the same analyses investigating the association between lifestyle, environmental and genetic factors with those with IBS and those on a gluten-free diet. The full cohort (approx. 500,000 participants) will be included.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-epidemiological-study-of-genetic-and-clinical-factors-affecting-the-progression-of-age-related-macular-degeneration-amd

An Epidemiological Study of Genetic and Clinical Factors Affecting the Progression of Age-related Macular Degeneration (AMD)

Last updated:
ID:
58263
Start date:
2 March 2020
Project status:
Current
Principal investigator:
Dr Marcel van der Brug
Lead institution:
Clover Therapeutics Company, United States of America

Age-related macular degeneration (AMD) is a common eye condition and a leading cause of vision loss among people age 50 and older. If you have AMD, you might see a blurred area near your center of vision. Over time, the blurred area may grow bigger or you may have blank spots in your vision.
The purpose of this research is to discover causes of Age-Related Macular degeneration. Some of these clues are found in your DNA. DNA makes up your genes. Genes provide instructions for things like eye or hair color, height, and sometimes things that affect health. Everyone’s DNA is slightly different. By studying the DNA of many different people and their health records we hope to find differences that help people stay healthy or in some cases get sick with diseases like AMD.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-epidemiological-study-on-the-interplay-of-genetics-environment-and-biomarkers-in-liver-and-metabolic-health

An epidemiological study on the interplay of genetics, environment, and biomarkers in liver and metabolic health.

Last updated:
ID:
986948
Start date:
20 August 2025
Project status:
Current
Principal investigator:
Mr Yang Qiu
Lead institution:
Longhua Hospital Shanghai University of Traditional Chinese Medicine, China

This project will investigate the interplay of genetics, lifestyle, and biomarkers in the pathogenesis of chronic liver diseases (e.g., fatty liver, cancer) and related metabolic disorders. Using the UK Biobank’s multi-modal data, we aim to elucidate aetiological pathways to improve disease prediction and prevention.

Research Questions:
What are the key genetic and lifestyle risk factors for adverse liver outcomes?
How does genetic susceptibility interact with modifiable lifestyle factors to influence liver and metabolic disease risk?
Can multi-omic biomarkers improve the early detection and risk stratification of liver disease?
What are the causal relationships between metabolic traits and the risk of severe liver disease?

Objectives:
To quantify the effects of genetic and lifestyle factors on liver health outcomes.
To evaluate how genetic background modifies the impact of environmental exposures on disease risk.
To identify and validate biomarkers for early diagnosis and prediction of disease progression.
To develop integrative models combining multi-modal data for improved risk prediction.

Scientific Rationale:
Chronic liver disease and associated metabolic dysfunction are a major public health burden. Their development is multifactorial, but the gene-environment interplay is poorly understood, hindering effective prevention. The UK Biobank’s large-scale, prospective design with deep genetic and phenotypic data offers an unparalleled resource to address this knowledge gap. This research will advance our understanding of disease mechanisms and provide an evidence base for novel prevention strategies and personalised risk assessment.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-examination-of-cardiometabolic-risk-and-event-rates-in-individuals-who-have-undergone-bariatric-surgery

An examination of cardiometabolic risk and event rates in individuals who have undergone bariatric surgery

Last updated:
ID:
97880
Start date:
7 September 2023
Project status:
Current
Principal investigator:
Dr Brian T Steffen
Lead institution:
University of Minnesota Twin Cities, United States of America

Obesity in industrialized nations is a deepening health crisis. Among the many strategies that have been developed to address it, weight loss surgery remains one of the most effective tools for sustained weight loss-particularly for those with severe obesity. Bariatric surgery (BarS) causes considerable weight loss, and patients have been shown to lose over 60% in excess bodyweight and 30% in total bodyweight depending on age, sex, and patient health. In addition, improvements in heart and metabolic health are typical. Following surgery, patients have been shown to have normal or reduced blood sugar, cholesterol levels, and blood pressure. Despite the evidence that BarS improves health outcomes over the short-term, few studies have examined long-term outcomes such as cardiovascular disease, diabetes, and mortality. In addition, there are few studies that have looked at what lifestyle factors, like diet, contribute to better outcomes in over the long-term. Finally, some researchers have proposed that, even when an individual does not lose a lot of weight following BarS, there will still be improvements in blood sugar and cholesterol. Altogether, more research is needed to examine the health outcomes of BarS over time.

The proposed research will compare health outcomes between UK Biobank participants who have undergone a BarS procedure with two groups of study participants: those who have not undergone one of these surgeries but are of the same age, sex, race/ethnicity and 1) have the same BMI (bodyweight in kg/height in meters2) as those who had a BarS procedure prior to their surgery; and 2) have the same BMI as those who had a BarS procedure after their weight loss surgery. We aim to examine whether there are health benefits to BarS that depend on the weight loss that is obtained (by comparing BarS patients with group 1) and there are health benefits to BarS that are independent of the weight loss that is obtained (by comparing BarS patients with group 2). Finally, the research aims to determine the lifestyle and other factors that contribute to better and worse health outcomes following BarS over the long-term.

This research is part of a grant application to the National Institutes of Health, and will be conducted over the course of four years. This research will describe the health outcomes of BarS patients and the lifestyle factors that lead to better or worse outcomes.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-examination-of-the-association-between-germline-genetic-polymorphisms-and-urinary-bladder-cancer-survival

An examination of the association between germline genetic polymorphisms and urinary bladder cancer survival

Last updated:
ID:
53618
Start date:
16 October 2019
Project status:
Closed
Principal investigator:
Dr Michael Passarelli
Lead institution:
Dartmouth College, United States of America

Despite recent therapeutic advancements, urinary bladder cancer remains the most expensive cancer to treat. Previous studies have discovered germline genetic variants that increase risk for urinary bladder cancer, however little is known about whether these variants are related to survival time after a diagnosis of bladder cancer. Here, we propose to use the genome-wide genotyping data of individuals diagnosed with urinary bladder cancer in the UK Biobank to validate results from a genetic association study of bladder cancer-specific and overall survival conducted in the New England region of the United States. These findings may prompt future studies that help improve therapeutic decisions in the clinical setting.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-explainable-machine-learning-framework-for-early-stratification-of-cardiovascular-risk-in-type-2-diabetes

“An Explainable Machine Learning Framework for Early Stratification of Cardiovascular Risk in Type 2 Diabetes”

Last updated:
ID:
935792
Start date:
28 August 2025
Project status:
Current
Principal investigator:
Miss Anupama A. Pandey
Lead institution:
Delhi Technological University, India

My research aims to develop an explainable machine learning framework to support early identification of cardiovascular risk in individuals with Type 2 Diabetes Mellitus (T2DM). Cardiovascular disease is a major complication of T2DM, yet early prediction remains challenging with traditional methods. This project will explore the use of large-scale health data from UK Biobank to build models that identify high-risk individuals, compare their performance with conventional risk scores, and apply explainable AI techniques (e.g., SHAP) to ensure transparency. The goal is to enable more accurate, timely, and personalized interventions, ultimately reducing preventable complications and improving long-term outcomes for people living with diabetes.

As part of this project, we are committed to responsible use of AI and ensuring that our research benefits the wider scientific and healthcare community. The outcomes of the study will be shared through peer-reviewed journal articles, conference presentations, and academic workshops.
To support transparency and reproducibility, we also plan to share relevant code, analysis pipelines, and trained models via trusted open platforms like GitHub-wherever it is permissible under UK Biobank’s data sharing policies. this contains how many character
No individual-level data from UK Biobank will be made public at any stage. All dissemination efforts will strictly follow the UK Biobank AI publication guidelines, ensuring ethical research conduct and meaningful public benefit.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-exploration-of-how-the-neighbourhood-environment-personal-characteristics-and-genetic-variations-contribute-to-geographical-variation-in-the-prevalence-of-diabetes-and-obesity

An exploration of how the neighbourhood environment, personal characteristics and genetic variations contribute to geographical variation in the prevalence of diabetes and obesity

Last updated:
ID:
58421
Start date:
30 March 2020
Project status:
Closed
Principal investigator:
Miss Yunqi Zhou
Lead institution:
University of Bristol, Great Britain

The causes of chronic diseases such as diabetes and obesity have been widely studied and related to various factors including individual behaviours and genetics, as well as to the neighbourhood environments in which people live. Less studied is the possibility that these causes and their effects are themselves modified according to a wider geographical context, creating geographical variations in health outcomes and their causes.

This study will adopt methods of spatial analysis and of geographical modelling to explore geographical variations in the prevalence of diabetes and obesity, and whether their causes vary from places to places. It aims to understand if and how geographical context plays an important role in generating health disparities and to examine the potentially geographically varying relationships between various types of risk factors (including the neighourhood environment, personal factors and genetic variations). It will consider how lifestyles, personal characteristics and neighbourhood environment attenuate or increase any genetic predisposition to chronic diseases and whether that attenuation or increase is more characteristic of some places more than others. Particular attention will be paid to how the neighbourhood environment, such as household deprivation interacts with genetic variations and how these interactions vary over space.

The fours aims of the study are:
Aim 1: To quantify and to visualize the geographical variation of two chronic diseases – diabetes and obesity
Aim 2: To explore the extent to which the geographical variation in the prevalence of these diseases can be explained by the neighbourhood (built environment) and lifestyle factors
Aim 3: To look at how and if genetic factors interact with environmental and personal lifestyle factors to explain the geographical variation
Aim 4: To investigate how the modelled relationships may themselves vary geographically; for example, does an unhealthy lifestyle matter more in one city compared with another?

This project duration is 36 months for the PhD thesis under the supervision of Professor Richard Harris and Dr Emmanouil Tranos. By better understanding the geographically varying correlates of diabetes and of obesity, the study can help inform better prevention strategies, especially for vulnerable groups and high-risk regions, avoiding “one-size-fit-all” interventions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-integrated-approach-to-understanding-the-complexity-of-brain-disorders-for-precision-medicine-development

An Integrated Approach to Understanding the Complexity of Brain Disorders for Precision Medicine Development

Last updated:
ID:
135200
Start date:
21 March 2024
Project status:
Current
Principal investigator:
Professor Koji Kamagata
Lead institution:
Juntendo University, Japan

Currently, most neurodegenerative and psychiatric disorders are mainly treated symptomatically as no fundamental cure has been established, but the effectiveness of this treatment is limited and varies greatly from person to person. This is due to the fact that the onset and progression of neurological diseases can be influenced not only by genetic factors but also by environmental factors. Furthermore, the mechanisms by which diseases known to adversely affect multiple organs including the brain, such as cancer, lifestyle-related diseases, and COVID-19, cause cognitive dysfunction, as well as their relationship to genetic and environmental factors, have not yet been fully investigated. To address this challenge, this project proposes an integrated approach to simulate the complexity of brain disorders and ultimately support the development of precision medicine.
The significance of this project is in accordance with the objectives of UK Biobank. In conclusion, this research project proposes an integrated approach to determine the complexities of brain disorders. By combining diverse data modalities, this research project aims to identify critical factors that influence disease development and determine disease comorbidity patterns. The findings of this study are expected to have significant implications for the advancement of precision medicine in the treatment of brain disorders. This study will be supported by the Japan Society for the Promotion of Science Grants-in-Aid for Scientific Research (KAKENHI; Grant numbers 23H02865). The research period is planned to be 36 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-integrated-biopsychosocial-approach-to-identify-shared-mechanisms-underlying-major-psychiatric-and-neurodegenerative-disorders

An integrated biopsychosocial approach to identify shared mechanisms underlying major psychiatric and neurodegenerative disorders

Last updated:
ID:
106223
Start date:
9 August 2023
Project status:
Current
Principal investigator:
Dr Gemma Sharp
Lead institution:
University of Exeter, Great Britain

Individuals diagnosed with psychiatric disorders, such as depression, anxiety, and post-traumatic stress disorder (PTSD), have an increased risk of developing neurodegenerative disorders like Alzheimer’s and Parkinson’s later in life. Likewise, individuals with neurodegenerative diseases are at a higher risk of experiencing psychiatric symptoms. These findings suggest shared underlying mechanisms between these conditions. Understanding these shared mechanisms can inform the development of strategies to predict, prevent, and treat both psychiatric and neurodegenerative disorders. Additionally, intervening in psychiatric disorders earlier in life may have the added benefit of preventing neurodegenerative disorders later on.

Current research primarily focuses on biological factors to find potential drug targets, but mental health and brain diseases are influenced by a mix of biological, psychological, and social/environmental factors. Therefore, we propose an integrated biopsychosocial approach to identify shared mechanisms between these disorders. Our research aims to explore associations between common mental health and neurodegenerative disorders, examining biological measures (genetics, proteomics, metabolomics) and environmental/psychosocial factors (diet, personality, living arrangements, occupation) throughout life. By examining evidence from different angles, we hope to understand how these factors work together to contribute to the development of these conditions.

To find answers, we will use different methods. We will look at a large amount of data to see if certain factors are related to mental health and brain diseases (phenome-wide association studies). We will also use statistical analyses and advanced models to understand how these factors work together (regression analyses and structural equation modeling). By studying genes, proteins, brain scans, and other measures, we can learn more about the biological aspects. We will also explore if certain factors play a causal role in these conditions using innovative statistical techniques.

This research aims to enhance our understanding of shared and independent mechanisms underlying mental health and neurodegenerative disorders. It holds the potential to develop interventions targeting various aspects of physiology, behavior, environment, and social structure, with significant implications for public health. Ultimately, the findings can improve prediction, prevention, and treatment strategies for both sets of conditions and contribute to a better understanding of resilience.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-integrated-exploration-of-behavioral-environmental-and-genetic-predictors-associated-with-the-onset-and-prognosis-of-cardiovascular-disease-digestive-diseases-and-multimorbidity

An Integrated Exploration of Behavioral, Environmental, and Genetic Predictors Associated with the Onset and Prognosis of Cardiovascular Disease, Digestive Diseases, and Multimorbidity

Last updated:
ID:
564547
Start date:
9 January 2025
Project status:
Current
Principal investigator:
Dr Duguang Li
Lead institution:
Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, China

Modifiable and genetic determinants could jointly influence the onset and development of cardiovascular and digestive diseases, as well as multimorbidity. However, research in this domain remains nascent. We aim to:
(1) identify modifiable risk factors and unrecognized biomarkers to elucidate their direct effects and potential moderating functions on gastro-cardiovascular disorders and co-morbidities;
(2) infer potential causal associations between different variables and gastro-cardiovascular disorders and co-morbidities;
(3) unravel genetic variants and candidate genes associated with higher disease susceptibility, and the level of proteins, metabolites and gene expression;
(4) discover potential genetic correlations between gastro-cardiovascular disorders and co-morbidities;
(5) provide mechanistic insight into how candidate genes exert their effects on disease development;
(6) identify potential molecular targets for drug repurposing.
(7) construct robust prognostic algorithms to predict the probability of disease onset, disease progression, life expectancy and incidence of other health-related endpoints for the early detection of high-risk populations.

Scientific rationale: Prospective cohorts yield extensive data for disease analysis and prevention. Genomic studies pinpoint disease-associated genetic variants and genes. Statistical and machine learning models enable risk factor screening, mediation analysis, effect size estimation, causal inference, and personal risk scoring. Single-cell RNA-seq integration reveals gene roles in disease pathology, informing modeling and drug targets. Transcriptome-wide association study elucidates gene expression regulation in diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-integrated-exploration-of-behavioral-environmental-social-economic-genetic-microecological-predictors-associated-with-the-onset-and-prognosis-of-respiratory-diseases-and-multimorbidity

An Integrated Exploration of Behavioral, Environmental, Social economic, Genetic, Microecological Predictors Associated with the Onset and Prognosis of Respiratory diseases and Multimorbidity

Last updated:
ID:
688295
Start date:
24 February 2025
Project status:
Current
Principal investigator:
Professor Wentao Ni
Lead institution:
Peking University People's Hospital., China

Respiratory diseases are leading causes of global morbidity and mortality, with complex pathogenesis involving genetic, environmental, behavioral, and socioeconomic factors. Respiratory diseases are intricately linked to multiple organ systems, including cardiovascular, digestive, neuropsychiatric, musculoskeletal, urogenital, immune, hematologic, and endocrine systems, et al. The development of respiratory diseases is multifactorial, influenced by genetic predispositions, environmental exposures, behavioral factors, socioeconomic status, and microbial imbalances.
Large-scale cohorts like the UK Biank provide long-term data to study these interactions. Genomic studies (GWAS, QTL) identify genetic loci and candidate genes, while single-cell RNA-seq reveals cell-type-specific molecular mechanisms. Transcriptome-wide association studies (TWAS) and multimodal research elucidate gene expression regulation. Machine learning models enable risk factor screening, causal inference, and personalized risk prediction, offering tools for precision prevention and treatment.
Research Questions
Which behavioral, environmental, socioeconomic, genetic (multi-omics), and microecological factors predict respiratory disease onset and prognosis?
How do these factors interact to influence disease progression and complications?
How do respiratory diseases interact with other organ systems to drive multimorbidity?
What modifiable and genetic determinants jointly influence respiratory diseases and comorbidities?
Research Objectives: Identify predictive factors, Elucidate molecular mechanisms, Assess multimorbidity by exploring systemic associations, Infer causal relationships, Unravel genetic variants, Discover genetic correlations between respiratory diseases and comorbidities, Identify drug targets for repurposing, Develop predictive models.
This study aims to unravel the complexity of respiratory diseases and their systemic interactions, advancing precision medicine.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-integrated-genetic-neuroimaging-and-behavioral-understanding-of-sleep

An integrated genetic, neuroimaging, and behavioral understanding of sleep

Last updated:
ID:
465546
Start date:
28 November 2024
Project status:
Current
Principal investigator:
Professor Chen-Ying Huang
Lead institution:
National Taiwan University, Taiwan, Province of China

Behavioral changes and brain activity alterations can provide important information about potential diseases, allowing for earlier intervention and better management of conditions. Sleep plays a crucial role in various physiological and psychological processes. We intend to use sleep as a starting point to conduct integrated research.

The over-arching goal of this study is to elucidate the neural network subserving sleep and identify its genetic origins and behavioral manifestations. Specific aims include: 1) identifying the functional connectivity related to the suprathalamic nucleus (SCN), a part of basal ganglia supporting circadian rhythm regulation, 2) revealing how the SCN network is related to sleep behaviors, including subjective and objective measures, 3) discovering the genetic underpinning accounting for the variability of sleep behaviors across individuals, and 4) investigating how SCN-network connectivity mediates the genetic effect on sleep.

Sleep serves as a starting point. With these understandings, we will extend the study goals toward other interactions between neural and genetic mechanisms underlying various phenotypes and quantitative traits, especially those related to behaviors. UK Biobank has an extensive collection of behavioral measurements other than sleep. We are particularly interested in mental health, well-being, and cognitive functions. Sleep can heavily influence these behaviors. We aim to see how sleep mediates the genetic and neural effects on these behaviors. We summarize this relationship as gene-brain-sleep-behavior.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-integrated-investigation-of-genetic-behavioral-environmental-socioeconomic-metabolic-and-microbiome-predictors-influencing-the-onset-progression-and-multimorbidity-of-diabetes

An Integrated Investigation of Genetic, Behavioral, Environmental, Socioeconomic, Metabolic, and Microbiome Predictors Influencing the Onset, Progression, and Multimorbidity of Diabetes

Last updated:
ID:
836469
Start date:
30 July 2025
Project status:
Current
Principal investigator:
Ms Xiaobin Lin
Lead institution:
Second Xiangya Hospital of Central South University, China

Diabetes is a major global health concern, with rising prevalence linked to significant morbidity and mortality. This project aims to dissect the multifactorial nature of diabetes by examining the interplay of genetic predisposition, lifestyle factors (such as diet and physical activity), socioeconomic influences, and the gut microbiome in the onset and progression of diabetes, including its multimorbid consequences.
Key research questions include:
1. What genetic variants are associated with increased diabetes susceptibility in the UK Biobank cohort?
2. How do lifestyle and behavioral factors interact with genetic risk to influence diabetes development and outcomes?
3. What role do socioeconomic factors and environmental exposures play in diabetes management and health disparities?
4. How does the gut microbiome contribute to metabolic changes associated with diabetes?
The objectives of this study are to:
1. Identify genetic loci associated with diabetes and related conditions through genome-wide association studies (GWAS).
2. Assess the impact of dietary habits and physical activity on diabetes risk.
3. Investigate environmental influences, such as air pollution and socioeconomic status, on health outcomes.
4. Analyze the role of microbiome diversity in metabolic health and diabetes progression.
Utilizing the extensive dataset from the UK Biobank, this research will provide critical insights into the complex relationships among these variables, contributing to the development of personalized prevention and treatment strategies for diabetes. Ultimately, this project aims to improve health outcomes and clinical management for individuals with diabetes and multimorbidity.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-integrated-multi-omics-study-of-novel-biomarkers-in-aging-and-aging-related-diseases

An Integrated Multi-Omics Study of Novel Biomarkers in Aging and Aging-Related Diseases

Last updated:
ID:
262935
Start date:
13 January 2025
Project status:
Current
Principal investigator:
Dr Peilin Jia
Lead institution:
Beijing Institute of Genomics, China

aims
Our research aims to explore the influence of multiple omics features (genomics, proteomics, metabolomics, and exposome, etc.) and their intricate interactions on aging and aging-related diseases, providing new perspectives for deciphering biological processes and molecular events. Furthermore, by pinpointing shared signals between molecular phenotypes and disease GWAS, we aim to unveil the black box from genetic variation to the onset of diseases, understanding the underlying biological mechanisms.

scientific rationale
Currently, the global population of elderly people is growing at an unprecedented rate. It is projected that by 2050, the population aged 65 and above will reach 1.6 billion. The continuous increase in incidence of age-driven chronic diseases will place a substantial burden on global socioeconomic frameworks. In this context, the scientific challenge of how to address aging, with the goal of fostering healthy aging, has come into the limelight.

project duration
This project is expected to be completed within 36 months.

public health impact
The results of this research will contribute to a comprehensive understanding of the developmental process of chronic diseases and support precise personal prevention. Concurrently, the findings will provide invaluable evidence for the general public and organizations, enhancing their understanding of public physical and mental health. This will assist in improving lifestyle choices, preventing chronic diseases, and enabling the adoption of early disease prevention interventions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-integrated-pharmacogenomic-study-drug-safety-efficacy-and-the-role-of-gene-enviornment-interactions

An integrated pharmacogenomic study: drug safety, efficacy and the role of gene-enviornment interactions.

Last updated:
ID:
879030
Start date:
5 August 2025
Project status:
Current
Principal investigator:
Dr Malgorzata Maria Borczyk
Lead institution:
Maj Institute of Pharmacology of the Polish Academy of Sciences, Poland

SCIENTIFIC RATIONALE: Large-scale evidence shows that pharmacogenetic (PGx) testing improves clinical outcomes [PMID:36739136]. However, many PGx variants and their interactions with other factors remain unknown [PMID:36707729]. Large-scale population biobanks coupled with electronic health records, including prescriptions, offer new possibilities for high-throughput PGx research [PMID:38633781]. We hypothesise that, among other traits, age and stress-related endocrine status are important mediators of drug response.

RESEARCH QUESTION: Which genetic variants and measured phenotypes associate with adverse reactions and therapeutic success?

AIMS AND OBJECTIVES: (1) To build a database of drug-response profiles from EHRs (e.g. adverse reactions, dose adjustments, therapy lengths, medication switches) for drugs acting on cardiovascular and nervous systems; (2) to build a database of glucocorticoid receptor (GR) activity/HPA axis-related phenotypes as readout of endocrine status (3) to perform genome-wide multi-trait association studies using novel methods [https://doi.org/10.1101/2025.04.29.25326633] to extract both genetic variants as well as other measured factors (e.g. age, other medication, GR/HPA phenotypes) associated with drug response phenotypes. We will include any relevant covariates (e.g., local population stratification).; (4) to build multifactor predictive models to predict drug response phenotypes from step 1; (4) to study in detail how age and HPA axis activation interact with PGx variants and how they contribute to prediction.

OUTCOMES: Our integrated approach should reveal novel markers and generate testable hypotheses for pharmacogenomics. Results will be published at 2+ international conferences and in 3+ peer-reviewed publications. All of the code generated will be released to the scientific community.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-integrative-approach-to-predict-mental-health-outcomes-the-contribution-of-genetics-brain-architecture-and-behavior

An Integrative Approach to Predict Mental Health Outcomes: The Contribution of Genetics, Brain Architecture, and Behavior

Last updated:
ID:
105731
Start date:
16 November 2023
Project status:
Current
Principal investigator:
Professor Giulio Pergola
Lead institution:
Lieber Institute for Brain Development, United States of America

Psychotic and mood disorders are complex and challenging to understand due to their varied symptoms and complex genetic architecture. However, large-scale datasets like the UK Biobank provide an excellent opportunity to study the genetic factors associated with these disorders. Previous research has shown that genetic risk in non-coding areas of the genome can influence gene expression across different brain regions, leading to observable effects on behavior and neurophysiological correlates.
This project, estimated to last three years, aims to build on that knowledge by assessing variability in structural and functional brain architecture using Magnetic Resonance Imaging (MRI)-derived estimates. The goal is to investigate how these estimates might predict mental health-related outcomes.
Considering the significant role of sociodemographic, psychosocial, cognitive, physical health, and lifestyle factors as risk factors for psychotic and mood disorders, this project aims to leverage all of these measures to develop a comprehensive understanding of the genetic contributions to these disorders.
Specifically, the project will focus on identifying gene co-expression pathways that underlie region-specific brain characteristics. By computing individual parsed polygenic co-expression indices, the project will explore the relationship between gene expression and structural and functional brain characteristics using multiple MRI modalities that capture interindividual brain variability. Additionally, the project will utilize supervised and unsupervised machine learning algorithms to define individual profiles based on the relationship between cognitive and gene-brain features. These profiles will then be used to predict mental health-related outcomes.
The findings of this project will be disseminated through open-access platforms, allowing for replication and dissemination of the results. The extensive and valuable data available in the UK Biobank makes it an ideal dataset for this project, which aims to contribute to the growing field of personalized medicine in mental health care.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-integrative-data-driven-approach-to-the-genomics-of-anti-tuberculosis-and-anti-malarial-drug-responses-in-african-populations

An integrative data-driven approach to the genomics of anti-tuberculosis and anti-malarial drug responses in African populations

Last updated:
ID:
96110
Start date:
7 March 2024
Project status:
Current
Principal investigator:
Dr Houcemeddine Othman
Lead institution:
Wits Health Consortium (Pty) Ltd, South Africa

Response to drugs is tightly controlled by a set of genes responsible for the absorption, distribution, metabolisation, and excretion of drugs in the organism (ADME genes). The genetic variability of ADME genes differs significantly among world populations. In particular, Africa, as a place of birth of the genus Homo, and given its variety of climates and evolutionary constraints, is the most diverse continent in the world. Therefore, population groups in Africa might respond differently to drugs, particularly those used to treat infections with malaria and tuberculosis. These two transmissible diseases are major health and economic burdens in Africa. Therefore, profiling genetic variability in Africa is a key step in understanding biological mechanisms that control response to anti-malaria and anti-tuberculosis drugs. High throughput Genomic technologies such as third-generation sequencing and microarray genotyping offer the opportunity to characterize the ensemble of ADME gene variability in batch. Uk Biobank contains a large repertoire of such data related to individuals from Africa or with African ancestry that can help in profiling the generic variability of genes involved in anti-malaria and anti-tuberculosis drug treatment. These data could be processed using computational methods to determine the variants with potentially significant impact on the function of ADME genes. With the help of UK Biobank data, we will study the impact of African genetic variability on drug processing mechanisms involving 35 drugs that are in use or with high pharmacological potential for the treatment of tuberculosis and malaria infections. The study is scheduled for two years period and will involve other datasets collected from different collaborators in addition to data from the UK Biobank. Our study will help in the efforts of establishing tailored therapies against malaria and tuberculosis to increase the efficacy of drugs in African population groups.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-integrative-multi-modal-investigation-into-the-mechanisms-trajectories-and-intervention-targets-of-affective-anxiety-and-trauma-related-disorders

An Integrative Multi-modal Investigation into the Mechanisms, Trajectories, and Intervention Targets of Affective, Anxiety, and Trauma-Related Disorders

Last updated:
ID:
925561
Start date:
4 September 2025
Project status:
Current
Principal investigator:
Dr Jingchu Hu
Lead institution:
Shenzhen Kangning Hospital, China

Scientific Rationale:
Affective, anxiety, and trauma-related disorders are leading causes of disability with complex, heterogeneous etiologies. Current research is often fragmented, failing to capture the interplay of risk factors over time. A critical gap exists in understanding the longitudinal trajectories of these conditions. This project will leverage the UK Biobank’s unique multimodal data to build an integrative biopsychosocial model, aiming to uncover the mechanisms that drive illness and resilience.

Research Questions:
1. How do genetic predispositions, environmental factors, and lifestyle behaviors (e.g., sleep, activity) interact to shape the developmental trajectories of these disorders?
2. What are the core biological mechanisms, including physiological and neurobiological pathways, that mediate these risk trajectories?
3. Can we synthesize these high-dimensional, multi-modal data to accurately predict an individual’s risk and identify novel, modifiable targets for intervention?

Objectives:
1. To Map Longitudinal Trajectories: We will integrate genetic data (Polygenic Risk Scores), health records, and objective lifestyle measures from accelerometry to identify and characterize distinct temporal patterns of risk and symptoms.
2. To Investigate Biological Mechanisms: We will analyze biochemical markers (from blood/urine assays) to probe the role of mediating physiological pathways (e.g., inflammation). This provides the foundation for future investigation into neurobiological substrates.
3. To Develop Predictive and Interventional Models: We will employ machine learning algorithms to build robust, integrative models that predict individual risk. The ultimate goal is to pinpoint the most impactful and modifiable targets for personalized interventions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-integrative-study-of-multi-dimensional-risk-factors-in-cardiovascular-diseases-physiological-environmental-behavioral-genetic-and-socioeconomic-perspectives

An integrative study of multi-dimensional risk factors in cardiovascular diseases: Physiological, environmental, behavioral, genetic, and socioeconomic perspectives

Last updated:
ID:
920292
Start date:
17 July 2025
Project status:
Current
Principal investigator:
Dr Manqi Zheng
Lead institution:
Capital Medical University, China

Cardiovascular diseases (CVDs)-encompassing heart and blood vessel disorders -remained the leading global cause of mortality, accounting for 19.4 million deaths (28.6% of all fatalities) in 2021. A paradox persists: while WHO notes most CVDs are preventable via behavioral/environmental risk factors, global burden continues rising, highlighting urgent need to enhance prevention strategies.
Current evidence emphasizes multi-domain interventions targeting modifiable factors (amenable to lifestyle/behavioral/policy-driven societal changes). However, critical gaps persist: prior studies focused narrowly on hypothesized risk factors under hypothesis-driven frameworks, potentially overlooking unhypothesized contributors; risk factors often manifest as interconnected clusters; and univariate significant factors may lose robustness in multivariate contexts. Consequently, such approaches fail to capture CVD’s multifactorial etiology and risk factor synergies, exacerbating selective reporting and publication bias. Besides, non-standardized analytical methods introduce heterogeneity, hindering cross-study comparability.
To bridge these gaps, this project aims to 1) systematically identify multi-dimensional modifiable CVD risk factors (physiological/environmental/behavioral/socioeconomic) using UK Biobank’s large-scale prospective cohort; 2) examine interactions between modifiable exposures and genetic variations in CVD risk; and 3) quantify synergistic effects on CVD incidence and evaluate the public health impact of multi-domain interventions.
Our dissemination strategy encompasses academic publication in high-impact journals and presentation at international conferences to engage the scientific community. Simultaneously, we will develop accessible public science materials and community health education campaigns to translate findings into actionable prevention strategies, ensuring our research directly informs both clinical practice and population-level health policies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-international-collaboration-to-identify-the-genetic-variants-of-diabetic-retinopathy

An international collaboration to identify the genetic variants of diabetic retinopathy

Last updated:
ID:
50604
Start date:
29 August 2019
Project status:
Current
Principal investigator:
Dr Weihua Meng
Lead institution:
University of Nottingham Ningbo Campus, China

Diabetic retinopathy (DR) is the most common eye complication in diabetic patients and the most common cause of blindness among people of working age in the UK. Around 30% type 2 diabetes will develop DR and this represents 1.5 million people in the UK. The quality of life for DR patients can be significantly affected due to visual impairment, worries and movement restrictions. In addition to physical and emotional impacts, DR also represents a significant economic burden to the healthcare system. Therefore it is important to study the mechanism of DR which might lead to a method to reduce the impact of this disorder.
The genetic mechanism of DR is far from being understood despite previous studies have confirmed that DR is a disorder with genetic components. There have been some genome-wide association studies on DR while the results need further confirmation.
The UK Biobank has recruited 30,000 participants with diabetes and among them, over 2,200 have developed DR. This allows us to perform a standard genome-wide association study on DR aiming to identify the genetic components. In addition, we will perform a GWAS meta-analysis using the GWAS summary statistics on DR provided by researchers of the International Diabetic Retinopathy Consortium on Genetics (IDRCG). Our study combining UK Biobank and IDRCG will be by far the largest one on DR.
The project will last around 1.5 years. We will find the genetic variants for diabetic retinopathy which will fill the gap in the current understanding of this order. The genetic finding of our project might imitate the development of effective treatments such as identifying genetic variants with potential to be a drug target for prevention or treatments.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-investigation-into-the-genetic-overlap-between-anxiety-disorders-psychiatric-comorbidities-and-neuroimaging-phenotypes

An Investigation into the Genetic Overlap between Anxiety Disorders, Psychiatric Comorbidities and Neuroimaging Phenotypes

Last updated:
ID:
71843
Start date:
14 February 2022
Project status:
Current
Principal investigator:
Professor Dan Joseph Stein
Lead institution:
University of Cape Town, South Africa

This proposal seeks to apply UK Biobank data to study the genetic relationship of human traits and altered brain structures. We aim to assess the genetic overlap between psychiatric disorders and associated health outcomes. Previous studies have successfully identified many genetic regions associated with psychiatric disorders. However, the genetic basis of many of these disorders is still largely unknown. We hypothesis that the genetics of these disorders remains undetected due to traditional statistical methods and that by leveraging novel statistical techniques we will be able to uncover the genetic overlap between traits. Uncovering the shared genetic causes of psychiatric disorders and altered brain structures could improve diagnosis, treatment outcomes and increase quality of life for patients. Thus, this proposal aligns with the stated aim of the UK Biobank ‘to improve the prevention, diagnosis and treatment of illness and the promotion of health throughout society’.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-investigation-of-anthropometry-physical-inactivity-metabolic-syndrome-and-type-2-diabetes-on-colorectal-cancer-development

An investigation of anthropometry, physical inactivity, metabolic syndrome and type-2 diabetes on colorectal cancer development.

Last updated:
ID:
25897
Start date:
2 February 2015
Project status:
Closed
Principal investigator:
Dr Neil Murphy
Lead institution:
International Agency for Research on Cancer, France

Obesity, physical inactivity, metabolic syndrome (metS) and type-2 diabetes (T2DM) have all been associated with higher colorectal cancer risk. However, previous studies investigating these relationships have usually been of smaller size, and many uncertainties remain, such as what are the biological pathways which link them. In phase 1 of the study, we will quantify the relationships between obesity, physical inactivity, metS and T2DM with colorectal cancer risk. In phase 2, we will use metabolic and hormonal biomarkers measured in all UK Biobank participants to investigate the possible biological pathways through which these disorders are linked. The planned analyses will aid the understanding of the obesity/inactivity/metS/T2DM and CRC relationships. The use of the metabolic and endocrinological biomarkers will highlight mechanisms through which these factors exert an effect on colorectal carcinogenesis. Presently, these molecular pathways are poorly understood with smaller previous studies yielding inconsistent results. Elucidation of these molecular pathways may aid risk prediction and identify possible therapeutic targets for CRC. In the full cohort of 500,000 men and women, information collected from participants at the onset of the study on measurements of obesity, physical activity levels, metS, and T2DM status will be used to assess the relationships between these factors and subsequent CRC risk.
Additionally, the relationships between the metabolic and hormonal biomarkers and metS, T2DM, obesity and physical activity, as well as CRC, will be assessed to shed light on the possible cancer pathways which these risk factors exert an effect. The analyses of anthropometry, physical activity, metS, and T2DM and CRC risk will be undertaken on the full cohort (phase 1). Similarly, when available in 2015, the relationships between the panel of measured biomarkers and CRC will be undertaken on the full cohort (phase 2). Mediation and pathway analyses will be undertaken at this stage to elucidate the molecular pathways which link metS, T2DM, obesity, physical inactivity with CRC risk. This proposed work will build upon previous analyses undertaken by members of our research group in the European Prospective Investigation into Cancer and Nutrition (EPIC) and the Women?s Health Initiative (WHI) studies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-investigation-of-auditory-processes-in-parkinsonism-disorders-risk-factors

An Investigation of Auditory Processes in Parkinsonism Disorders: Risk factors.

Last updated:
ID:
98097
Start date:
29 March 2023
Project status:
Current
Principal investigator:
Dr Megan Rose Hope Readman
Lead institution:
Lancaster University, Great Britain

Over 150,000 UK citizens have an incurable age-related brain disorder known as a neurodegenerative Parkinsonism disorder. Parkinson’s disease (PD) Progressive Supranuclear Palsy (PSP) and Multiple Systems Atrophy (MSA) are all types of neurodegenerative Parkinsonism disorders. These disorders are incurable; therefore, the identification of factors that increase the likelihood of disorder onset, and the occurrence of specific symptoms is vital.

Hearing loss (HL) increases the likelihood of dementia. This may be due to the abnormal accumulation of highly reactive molecules or alterations in specific proteins, which occurs in dementia, leading to the death of cells within the ear that are needed for hearing. Importantly, both of these processes occur in PD and MSA. However, whilst the accumulation of highly reactive molecules does occur, specific protein function is not altered in PSP. Therefore, HL may increase the likelihood of onset of some, but not all, Parkinsonism disorders.

Many individuals with Parkinsonism disorders experience cognitive difficulties (e.g. memory loss) and depression, however, not all do. HL may cause dementia and depression. Therefore, individuals with Parkinsonism disorders who also have HL may be more likely to experience cognitive decline and depression.

Demographic (e.g. sex and ethnicity) and socioeconomic (e.g. wealth and educational attainment) factors appear to influence the likelihood of onset of PD and the relationship between HL and depression. Therefore, it may be that demographic and socioeconomic factors influence the onset of Parkinsonism disorders and the extent to which HL increases the likelihood of onset of Parkinsonism disorders.

Despite the importance of understanding the relationship between HL and Parkinsonism disorders, no prior research has been conducted in this area. Consequently, we will conduct a large-scale analysis, of UK Biobank data, examining the relationships between HL, Parkinsonism disorders, cognitive function, depression and demographic and socioeconomic factors. This will take approximately 3 years.

This project has societal implications. Specifically, following current HL management policy individuals with dementia are classified as at risk of HL and receive more frequent audiology assessments. Thus, evidence that HL influences Parkinsonism disorders onset and symptoms would suggest that individuals with these disorders should also be categorised as at risk and receive more frequent hearing assessments. Additionally, the management of HL reduces cognitive function and depressive symptoms. Therefore, if HL influences cognitive impairment and depression in Parkinsonism disorders, then HL management may be beneficial in the management of these symptoms within these disorders. Thus these findings may instigate clinical trials.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-investigation-of-phenotypes-associated-with-the-snp-rs118174674-affecting-loxhd1

An investigation of phenotypes associated with the SNP rs118174674 affecting LOXHD1.

Last updated:
ID:
240789
Start date:
30 October 2024
Project status:
Current
Principal investigator:
Professor Nicolas Grillet
Lead institution:
Stanford University, United States of America

Age-related hearing loss poses a significant health concern, impacting over 50% of the global population by the age of 75. Previous genetic investigations have identified more than 50 small DNA variations that influence the susceptibility to age-related hearing loss.
The objective of this study is to ascertain whether one of these identified DNA alterations has additional implications beyond predisposing individuals to age-related hearing loss, such as an elevated risk of experiencing falls.
Should this hypothesis be validated, such insights could serve as invaluable tools for identifying and informing individuals at risk of falling.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-investigation-of-risk-factors-genetics-radiomics-and-health-care-of-physical-and-mental-conditions

An investigation of risk factors, genetics, radiomics and health care of physical and mental conditions

Last updated:
ID:
93474
Start date:
31 May 2023
Project status:
Current
Principal investigator:
Dr Xiaomei Zhong
Lead institution:
The Affiliated Brain Hospital of Guangzhou Medical University, China

Health is not merely the absence of disease or infirmity, but a balanced status of physical and mental conditions. Nowadays, ongoing physical and mental health conditions have affected a growing population around the world. Multidimensional factors are responsible for the onset and development of physical and mental conditions, while the precise mechanisms remain unclear. Moreover, physical health has been cited as an important determinant of mental health, and mental disorders can also impose considerable changes of physical conditions. Therefore, it is of great importance to investigate the mechanisms of mental and physical conditions, as well as their bi-directional relationships, to formulate intervention strategies in advance.
The research aim of this project is:
1) to study the independent and joint effects of genetics, risk factors, medications and social-economic status on physical and mental conditions in the UK Biobank cohort.
2) to combine cross-sectional and longitudinal data from biological measures, metabolomics and radiomics to achieve a comprehensive understanding of their potential mechanisms.
3) to explore the onset, recurrence, recovery and manifestions of physical and mental conditions, as well as their interlay mode.
4) to identify the health care and social-behavioral services for population, to facilitate policy-making for physical and/or mental problems.
Considering the expected computational and scientific complexity of this project, we expect to complete it within 3 years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-investigation-of-the-gene-x-environment-interactions-for-diabetes-and-metabolic-traits

An investigation of the gene x environment interactions for diabetes and metabolic traits

Last updated:
ID:
86619
Start date:
27 October 2023
Project status:
Current
Principal investigator:
Dr Shareefa Dalvie
Lead institution:
University of Cape Town, South Africa

Non-communicable diseases (NCDs), such as diabetes, result in 71% of deaths globally. Metabolic traits such as high blood pressure, obesity, insulin resistance, and high cholesterol increase the risk for certain NCDs. Diabetes and these metabolic risk traits are influenced by genes and environmental factors such as diet, physical activity and psychological trauma. However, exactly which genes and how these interact with the various environmental/lifestyle factors are largely unknown. In this proposed study we will investigate whether there are statistically significant interactions between genetic risk scores (which is a sum of the genetic risk variants across the genome) for diabetes and each of the metabolic traits and environmental factors and whether this interaction increases an individual’s risk for developing these medical traits. In particular, we are interested in the interactive effects between psychological trauma such as intimate partner violence and childhood trauma and the respective genetic risk scores for each of the traits in different ancestry groups. The findings from this study will provide more insight into the biology underlying diabetes and other NCDs and will also provide new knowledge on the relationship between genetic factors and the environment in the development of these complex diseases. This, in turn, will allow for the development of better prevention and treatment strategies. This study proposal aligns with the aims of the UK Biobank which is ‘to improve the prevention, diagnosis and treatment of a wide range of illnesses and to promote health throughout society’. We anticipate that this study will run for approximately 3 years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-mri-based-technology-for-early-assessment-of-antidepressant-efficacy-in-depression

An MRI-based technology for early assessment of antidepressant efficacy in depression

Last updated:
ID:
65920
Start date:
11 January 2021
Project status:
Current
Principal investigator:
Dr Jinkyeong Sung
Lead institution:
VUNO Inc., Korea (South)

Major depression disorder (MDD) is a serious health problem causing emotional distress, functional impairment, health problems, and suicide. However, more than 50% of patients are unable to find a suitable drug that works. This situation increases the duration of disease and makes treatment more difficult for patients. Thus, identifying effect of gene-smoking interaction on lung function in several populations is remaining task. Therefore, it is crucial to quickly select a drug that works for people with major depression disorder.

From previous studies, we identified some brain regions which related to antidepressants response treatment. However, the areas of the brain known to be crucial for antidepressants response treatment were very diverse and consensus was not reached. Therefore, our aim was to identify consistent important brain regions affecting MDD treatments and we will use multi-modality MRI traits in UKbiobank cohort. Moreover, we will develop antidepressants response treatment prediction model for reducing the time to select suitable drugs for MDD patients.

We expect that our finding provides understanding of MDD, reduces the burden on MDD patients, and emphasizes the importance of personalized medicine. Our plan was that One years for data construction, preprocessing, and integrating for use as training images, and additional two years for the development and application of prediction models. This project is expected to lead to great benefits for social economy and social health.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-mri-research-on-the-neural-mechanism-of-cognitive-impairment-dietry-behavior-and-psychology-in-diabetes-and-its-potential-application-in-clinical-diagnostic-models

An MRI research on the neural mechanism of cognitive impairment, dietry behavior and psychology in diabetes and its potential application in clinical diagnostic models

Last updated:
ID:
627927
Start date:
21 April 2025
Project status:
Current
Principal investigator:
Dr Ying Yu
Lead institution:
Fourth Military Medical University, China

Cognitive impairment, abnormal eating behavior, and abnormal psychological status are common brain functional damages in patients with diabetes. These damages interact and promote each other, severely affecting the quality of life of patients, compliance with disease interventions, and treatment outcomes. As the main body of the modern medical, the biopsychosocial medical model emphasizes that medical staff should not only focus on the biological factors of the disease during diagnosis and treatment but also consider the of psychological and social factors on the disease. This allows for a comprehensive and effective understanding of the patient’s disease situation, planning of scientific and reasonable treatment schemes, promotion of the patient’s early recovery to a healthy state. The purpose of this study is to explore the neural mechanisms, interactions, and potential clinical applications of cognitive impairment, abnormal eating behavior, and psychological abnormalities in patients with diabetes based on the classic biopsychosocial medical model, multimodel MRI and bioinformatic analysis.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-observational-and-genetic-investigation-into-the-association-between-atopic-dermatitis-and-risk-of-cardiovascular-kidney-metabolic-syndrome

An observational and genetic investigation into the association between atopic dermatitis and risk of cardiovascular-kidney-metabolic syndrome

Last updated:
ID:
904812
Start date:
19 August 2025
Project status:
Current
Principal investigator:
Mr Mingyang Yuan
Lead institution:
Third People’s Hospital of Chengdu, China

Research question: To explore the causal relationship and molecular mechanism between atopic dermatitis (AD) and cardiovascular- kidney -metabolic syndrome (CKM). Research aims: To explore the potential causal relationships between AD and various components of CKM through observational phenome-wide association study (PheWAS), polygenic risk score (PRS), as well as one-sample mendelian randomization and two-sample mendelian randomization. To investigate the tissue specificity of AD and components of CKM, in order to reveal potential molecular correlations. Scientific Rationale: To address these challenges, we employ several advanced genetic and statistical approaches, each with distinct principles and applications: 1. Observational PheWAS is a method to systematically explore the association between genetic variation and multiple phenotypes. Unlike traditional GWAS, which focuses on a single disease, PheWAS utilizes standardized phenotypic classification systems (such as ICD coding) to detect pleiotropy at genetic loci across a wide range of phenotypes. 2. PRS is a quantitative indicator that predicts an individual’s susceptibility to complex traits or diseases by aggregating the effect sizes of multiple genetic variants. 3. One-sample MR uses genetic variation in the same population as an instrumental variable (IV) to directly estimate the causal relationship between exposure and outcome. 4. Two-sample MR estimates causal effects through two independent sets of GWAS data, and deals with instrumental variable heterogeneity or horizontal pleiotropy by inverse variance weighting (IVW), MR-Egger and other methods. 5. Stratified LD score regression (S-LDSC) is a widely used estimation method for heritability enrichment. It enables quantification of SNP-based heritability of AD and CKM in various cells and tissues by using Z-statistic and corresponding P-values derived from regression coefficients. 6. Multi-marker Analysis of GenoMic Annotation (MAGMA) maps.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/an-observational-exploration-of-various-medication-risk-scores-calculated-from-electronic-health-record-data-in-a-general-population-to-predict-medical-outcomes

An Observational Exploration of Various Medication Risk Scores Calculated from Electronic Health Record Data in a General Population to Predict Medical Outcomes.

Last updated:
ID:
61422
Start date:
25 January 2021
Project status:
Current
Principal investigator:
Dr Sweilem Baseem Sweilem Al Rihani
Lead institution:
Tabula Rasa HealthCare, Inc., United States of America

With advanced age, providing medical care can present challenges as these patients are at risk for multiple chronic diseases . Inappropriate response to drugs and drugs-related adverse events (ADE) are an increasingly important problem in healthcare. The elderly are at an increased risk of ADE due to multiple chronic conditions leading to the intake of several drugs at the same time.. It is recognized that a high proportion of ADE in the elderly may be preventable. Increasing identification and prediction of ADEs have the potential to reduce the burden associated with ADE, will promote patient safety in the elderly and reduce medical costs.
The aim of this study is to use previously validated algorithms considering drug information to identify subsets of the population included in the UKBioBank at risk for ADEs. We hypothesized that the risk stratification based on medication claims data could help identify patients at high risk of medication related problems, as well as provide insights on interventions that could be performed by pharmacists or other health professionals to prevent these ADEs. In addition, we will assess the effect of using multiple medications that could predispose patients to dementia or pneumonia The calculated medication risk score (MRS) will be updated and validated in a population from UK Biobank and the predictive capacity will be compared to other currently used risk scores by the medical community. An MRS tool aiming to better predict ADE and identifying factors may have a significant impact in clinical practice from a clinical and economic perspective.
Research has shown that an estimated 20 – 50% of adults in the elderly population are prescribed at least one medication with properties that increase their risk of ADEs such as pneumonia or dementia. We hypothesized that a newly developed MRS that takes into account the dose of drug administered would be better to predict the risk of ADEs associated with these drugs Other side effects associated with some drugs that will be monitored by this MRS include constipation, dry mouth, and dry eyes. We also propose that the newly developed MRS could predict the frequency of visits to the emergency room or hospitalization. Our project will calculate the MRS each year for each patient, using prescribed drug information Results will be compared with the previous MRS.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/anaesthesia-related-disease-mechanism-and-perioperative-risk-based-on-multimodal-data

Anaesthesia related disease mechanism and perioperative risk based on multimodal data

Last updated:
ID:
797257
Start date:
23 July 2025
Project status:
Current
Principal investigator:
Dr Qian Wang
Lead institution:
Chinese PLA General Hospital, China

This project aims to investigate the biological mechanisms and potential predictive biomarkers of anesthesia-related outcomes, including cognitive decline, delirium, and pain. Leveraging the extensive genomic, proteomic, metabolomic, neuroimaging, and electronic health record data available in the UK Biobank, the study will integrate multi-modal data to identify risk factors, explore causality, and develop predictive models for these neurological complications.
Research Questions:
1.What are the shared and distinct molecular (proteomic/metabolomic) and neuroimaging signatures associated with sleep, cognitive decline, delirium, and pain?
2.Do genetic variants contribute causally to these outcomes, and how do they interact with perioperative exposures?
3.Can integrative models combining genetic, molecular, imaging, and clinical data accurately predict individuals at risk?
Objectives:
1.Use UK Biobank proteomic and metabolomic data to identify molecular pathways linked to adverse outcomes.
2.Analyze brain MRI to detect changes related to sleep, cognitive function and pain processing.
3.Apply Mendelian Randomization using genome-wide association data to test causal relationships between molecular traits and outcomes.
4.Develop machine learning-based predictive models that integrate multi-omics, imaging, and clinical data for personalized risk stratification.
Scientific Rationale:
Anesthesia and surgery may trigger long-term neurological effects, especially in genetically susceptible individuals. The UK Biobank provides a unique opportunity to explore these complex outcomes at scale, with operative health data, extensive omics datasets, and high-resolution brain imaging. Integrating these resources enables a systems-level understanding of how genetic and molecular profiles contribute to anesthesia-related cognitive and pain outcomes, paving the way for biomarker discovery and precision perioperative care.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analyses-of-genotype-to-phenotype-data-from-the-uk-biobank-to-aid-in-theidentification-and-validation-of-drug-targets

Analyses of genotype-to-phenotype data from the UK Biobank to aid in the identification and validation of drug targets

Last updated:
ID:
82416
Start date:
8 February 2022
Project status:
Current
Principal investigator:
Mr Pidong Li
Lead institution:
BioMap (Beijing) Intelligence Technology Ltd., China

The study of human genetic variation is an emerging paradigm for anticipating target-related
disease-relevance, including safety and efficacy targets.
The prerequisite of drug development is to identify a disease-relevant target that is druggable. BioMap endeavors to leverage multi-omic datasets using Artificial Intelligence (AI) technology to discover effective and safe drug targets or target combinations.
The proposed research projects aim to systematically perform association analysis between genetic variation and disease phenotypes to identify and validate potential drug targets in the disease areas, including but not limited to tumor immunology, autoimmune disease, and fibrosis. The UK BioBank dataset has genetic data for about 500,000 adults, along with extensive information about each individual. The combination of UK Biobank data and AI-based methodologies will enable us to develop efficient strategies to stratify patient subgroups better, identify and validate novel drug
targets, and assess benefits/risks associated with different patient characteristics.
Specifically, within BioMap’s research and development interests, we are focusing on developing therapeutics for
immune-related diseases, including but not limited to tumour immunology, autoimmune disease, and fibrosis. We anticipated the study would be completed in
around 36 months, with interest to be renewed afterward.
We hope the data offers opportunities for understanding disease biology, identifying and prosecuting novel drug targets, deriving strong evidence for patient stratification in clinical trials, and ultimately, contributing to unmet medical needs and significantly improving public healthcare.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analyses-of-il12b-in-complex-genetic-diseases

Analyses of IL12B in complex genetic diseases

Last updated:
ID:
95599
Start date:
9 March 2023
Project status:
Closed
Principal investigator:
Professor Grant Morahan
Lead institution:
Harry Perkins Institute of Medical Research, Australia

We have shown that a particular gene, IL12B, has variants associated with increased risk or severity of a number of diseases, including severe childhood asthma, fatal malaria, heart disease and cancer. This gene can be involved in many disease because it has the instructions to make an important protein that is involved in signalling and regulating the immune system, especially early in immune responses. However, the IL12B genetic variants are complicated and people can have one of eight different combinations; from our preliminary studies, each seeming to have different effects. These IL12B variants also are likely to interact differently with other genes to bring about disease. We would like to study and confirm the effects of the different Il12B variants in common diseases. We would also like to identify other genes that IL12B interact with.
Testing hundreds of thousands of genetic markers in patients or from the general population has allowed the identification of disease genes. Such genome-wide association studies have increased our understanding of the basis for many common diseases. However, most of the genes that have been identified contribute only a small amount of the overall risk of disease. Interactions of these genes can increase their impact. There are several methods to test for such interactions. These analyses are challenging because millions of independent interactions have to be calculated over a sample of thousands of patients.
The UK BioBank offers a great opportunity to confirm the role of IL12B in common diseases, and to identify other genes it interacts with. Production of genetic tests that can predict disease risk will allow earlier identification of people at risk of these diseases, allowing earlier intervention and better management. Prevention of CVD, DKD and cancers like melanoma will be very significant for patients and their families, and has the potential for enormous savings for the national health budget.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analyses-of-systemic-ageing-biomarkers-related-to-retina-via-deep-learning

Analyses of systemic ageing biomarkers related to retina via deep learning

Last updated:
ID:
68428
Start date:
20 July 2021
Project status:
Current
Principal investigator:
Dr Tyler Hyungtaek Rim
Lead institution:
Medi-Whale Inc., Korea (South)

Aims: The purpose of this study is to develop a novel biomarker that predicts biological age using retina, which is the layer of nerve cells lining the back wall inside the eye via Artificial Intelligence (AI), which refers to the simulation of human intelligence in machines that are programmed to think like humans and mimic their actions.

Scientific rationale: The blood vessels in the eye are where the body can observe the blood vessels directly. Many previous studies have shown that abnormal findings in the blood vessels or nerves in the eye are associated with systemic diseases or conditions. Recent advances in AI provide opportunities to further refine this area of research. In this study, we develop an AI system that can predict biological age by analyzing retinal photographs. UK Biobank is an important data for the validation of this AI system.

Project duration: 36months

Public health impact: Findings from this project may provide important information on biological ageing, which may improve our understanding of the complex mechanism involved in disease development or progression, and accelerate the development of prevention or early detection program for the major life-threatening disease. This project will also provide useful information on the generalizability of AI algorithms with the impact of genetically different populations.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysing-the-effects-of-privacy-protection-methods-on-data-utility-and-data-research-generalisability-in-observational-studies-a-case-study-in-asthma

Analysing the effects of privacy protection methods on data utility and data research generalisability in observational studies: A case study in asthma

Last updated:
ID:
49708
Start date:
18 June 2019
Project status:
Closed
Principal investigator:
Dr Mehrdad A Mizani
Lead institution:
University College London, Great Britain

Health data is the raw material to conduct data-driven observational studies. The results of these studies guide new clinical processes and policy makings for public health. However, releasing health data for research poses a risk to the privacy of individuals. Data holders remove data elements that directly identify individuals, such as name and personal information. However, this measure is inadequate by itself to protect privacy because a combination of remaining attributes (e.g. age, sex, occupation) can potentially leak the identity. Other measures are applied to protect privacy further. Here are some of the examples:
1) Removing the column on smoking habits
2) Deleting the record of patients older than 80
3) Aggregating data by only providing the average value of blood pressure for males and females of all ages.
5) The data is manipulated so that people with similar characteristics are put in groups with a minimum size, for example, a group of at least ten males 20-30 of age and another group for females.
In all these examples, the amount of precise information for research is reduced. For example, removing the smoking column would render the data less useful for finding a correlation between the disease and smoking.
Another issue is that applied manipulations on data to protect privacy are not informed of the researcher. Hence, the researchers will have uncertainties about the similarity of anonymised data to the original data in terms of relevant information to their research. This will lead to biased research results which in the long term will affect data-driven medical decisions and policy makings in public health.
This research aims to acquire anonymised asthma data from Biobank and further apply techniques used for anonymisation (e.g. deleting columns, aggregating values, creating synthetic records). This data will be used to analyse the effects of the anonymisation on the accuracy of observational methods in asthma. The research also aims at developing methods for measuring and characterisation of bias and loss of data usefulness. We will further investigate whether informing the researcher about the bias introduced by anonymity would improve the quality of research.
It should be emphasised that the aim of the research is not to re-identify data subjects or analyse the strength of the anonymity of acquired dataset.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysing-the-evolution-of-codon-usage-in-protein-coding-genes-from-proteome-data-of-54k-uk-biobank-participants-for-vaccine-design

Analysing the evolution of codon usage in protein-coding genes from proteome data of 54K UK Biobank participants for vaccine design

Last updated:
ID:
372175
Start date:
28 November 2024
Project status:
Current
Principal investigator:
Mrs Le Bao Xuyen Nguyen
Lead institution:
Hanyang University, Korea (South)

The COVID-19 pandemic has underscored the immense potential of messenger RNA (mRNA) vaccines as an innovative strategy for preventing infectious diseases. Despite their demonstrated advantages, mRNA vaccines encounter significant challenges, including instability, degradation, and inefficiencies in protein translation. To mitigate these issues, it is crucial to optimize the structure and codon usage of mRNA vaccine molecules. We propose to modify the mRNA sequence using a novel metric focused on the evolution of codon usage.
The primary objectives of this research are twofold. First, we aim to determine the relationship between point mutations and protein expression levels, as well as protein folding dynamics, by utilizing proteomics data. This will involve identifying the selective tendency of codon usage and correlating specific mutations with protein expression levels. Second, building on the findings regarding the selective tendency of human codon usage and existing indices, we intend to develop and validate a deep learning model. This model will serve as a sequence optimization tool for mRNA vaccine design, enhancing both translation elongation and protein structures as well as their functions.
To achieve these objectives, we plan to utilize the UK Biobank’s proteomics data to analyse the impact of point mutations on protein expression and folding. This analysis will focus on identifying patterns of codon usage and their effects on protein synthesis. We expect this study to provide valuable insights into codon usage bias across diverse organisms, thereby extending current knowledge in this area. Moreover, it is anticipated that our approach will overcome critical limitations in mRNA vaccine technology and significantly contribute to global health advancements.
We believe that our research has the potential to make substantial contributions to the field of mRNA vaccine development. By leveraging the comprehensive proteomics data available in the UK Biobank, we aim to pave the way for more effective and reliable mRNA-based therapeutics.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysing-the-relationship-between-cognition-and-image-based-phenotypes-through-explainable-ai

Analysing the relationship between cognition and image-based phenotypes through Explainable AI

Last updated:
ID:
106441
Start date:
16 April 2024
Project status:
Current
Principal investigator:
Dr Demian Wassermann
Lead institution:
Inria, France

This research project is using artificial intelligence to study the relationship between brain structure and function and cognition. The goal is to develop models that can accurately and specifically characterize these relationships. This could lead to new ways to diagnose, treat, and prevent cognitive disorders.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-and-computational-method-development-towards-unravelling-and-better-understanding-genetic-variation-underlying-cardiovascular-and-haematological-complex-traits

Analysis and computational method development towards unravelling and better understanding genetic variation underlying cardiovascular and haematological complex traits

Last updated:
ID:
34405
Start date:
14 September 2018
Project status:
Closed
Principal investigator:
Dr Valentina Iotchkova
Lead institution:
University of Oxford, Great Britain

The wealth of data and expertise for evaluating the genetic contribution to human traits in health and disease is continuously growing. However, the genetic architecture of complex traits, which are affected by multiple genes, lifestyle and environmental factors, still remains largely unexplained.

We propose to use the power of UK Biobank data, together with the development of novel computational methods to address the following key questions. First, to identify and understand genetic changes that lead to changes in phenotypic traits, such as haemoglobin levels or cardiovascular disease. Then, to use this information to infer direction of causality between different disease and disease risk factor traits. In parallel, we aim to identify currently unknown regulatory variants and regions with functional significance for gene regulation, human health and disease. Finally, we plan to explore the role of variability of the genomic sequence context and associated genetic variation in defining the initiation and progression of complex diseases.

Our main focus will be on quantitative measurements representing biomarkers of cardiovascular/haematological disease, however for comparison purposes, we plan to also examine different sets of traits, such as anthropometric measures.

Our research will expand basic science discoveries of human genetic variation through novel statistical and computational method development, and evaluation of whole-genome genotypes and cardiovascular and haematological traits. These efforts are expected to provide insights into the identification of new disease genes and druggable pathways. Additionally, interrogating regulatory variation in genomic regions, such as the globin clusters, has the potential to make a significant impact on global health as genetic variants in these regions influence anaemia and malaria with estimated 1.6 billion and 300-600 million people affected, respectively.

We request access to data on the full set of UK Biobank samples for a rolling 3-year period.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-alpha1-antitrypsin-related-comorbidities

Analysis of Alpha1-Antitrypsin related comorbidities

Last updated:
ID:
47527
Start date:
10 December 2019
Project status:
Closed
Principal investigator:
Professor Pavel Strnad
Lead institution:
Uniklinik RWTH Aachen, Germany

Severe Alpha1-antitrypsin deficiency (AATD) is the third most common genetic disorder leading to death and is caused mainly by the homozygous PiZ mutation (termed as Pi*ZZ). While lung and disease constitute the leading causes of AATD-related mortality, multiple other disorders including panniculitis and vasculitis have been described to associate with AATD.

While the Pi*ZZ genotype is relatively rare,the heterozygous carriage of PiZ allele (termed Pi*MZ) is seen in ~2% of European population, however the health risks associated with this genotype remain unknown. Recently, we demonstrated that PiZ carriage predisposes alcohol misusers as well as individuals with non-alcoholic fatty liver disease to development of liver cirrhosis. As the next step, we want to characterize the health risks associated with the Pi*MZ in the general population. For this purpose, we would like to use the UK biobank and compare a variety of metabolic and organ-related parameters between PiZ carriers and non-carriers. The parameters, that will display significant differences between the groups, will be subjected to multivariate analysis to account for confounders.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-biobank-neuro-imaging-data

Analysis of Biobank Neuro Imaging Data

Last updated:
ID:
8107
Start date:
1 October 2015
Project status:
Current
Principal investigator:
Professor Stephen Smith
Lead institution:
University of Oxford, Great Britain

We will apply image processing software to the neuro imaging data, in order to a) further develop image processing algorithms and software, for use by the wider research community (e.g. disseminating as part of our neuroimaging software FSL – FMRIB Software Library – www.fmrib.ox.ac.uk/fsl), and b) to co-analyse imaging-derived phenotypes against other Biobank data such as lifestyle measures and future health outcomes. The research will aid in extracting useful information (disease biomarkers, etc) from the Biobank imaging data. We will apply existing image processing software, and develop new software, for analysing the imaging data being acquired by Biobank. This will help us improve the software, and start extracting useful information from the data. All datasets where neuroimaging has been carried out.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-diabetes-mellitus-and-cardiometabolic-disease-in-relation-to-severe-outcomes-of-covid-19-a-uk-biobank-study

Analysis of diabetes mellitus and cardiometabolic disease in relation to severe outcomes of COVID-19: A UK Biobank study

Last updated:
ID:
62462
Start date:
28 August 2020
Project status:
Current
Principal investigator:
Dr Uazman Alam
Lead institution:
University of Liverpool, Great Britain

In this study, we will evaluate data from people with COVID-19 infection in relation to diabetes (type 1 and 2), high blood pressure, obesity and cardiovascular disease. We intend to study the characteristics of people with COVID-19 with particular reference to diabetes, high blood pressure, obesity and cardiovascular disease and the wealth of biomarker data in the UK Biobank. We will specifically investigate associations of interest based on the current understanding of people who are at risk. This includes clinical, biochemical and imaging biomarkers, background drug treatment and participants’ demographics such as ethnicity. We ultimately aim to develop a statistical model which will predict those who are at high risk of having adverse outcomes related to COVID-19 infection; resulting in the development of a robust tool that can be used for risk stratification of severe disease.

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a virus which results in COVID-19, primarily a respiratory illness. It is highly contagious and on 11th March the World Health Organisation labelled COVID-19 as a global pandemic. At time of writing, the number of deaths stands at more than 170,000 worldwide and unfortunately this figure is increasing exponentially. COVID-19 is a great threat to people’s physical, mental and economic well-being. According to available clinical data, up to 30% of the severe (requiring hospitalisation) COVID!19 patients have diabetes and hypertension and up to 15% have coronary heart disease. In addition, within the last week media reports suggest the majority of admissions to intensive care units in the UK for mechanical ventilation are people who are overweight or obese and Black, Asian and minority ethnic (BAME) people are disproportionately affected. Diabetes, cardiovascular disease and obesity seems to confer a much higher risk of severe COVID-19 than even chronic respiratory disease. It has also been suggested that diabetes medication including angiotensin converting enzyme inhibitors (ACEI), angiotensin receptor blockers (ARBs) and thiazolidinediones may result in increased susceptibility or severity of COVID-19 or are even beneficial.

If as expected this study demonstrates that people with diabetes and cardiovascular disease are at higher risk of severe COVID-19, these data will be utilised to develop a multi-state disease progression model for the prediction of disease severity and for the accurate identification/quantification of risk factors. Understanding the differences in risk of people with diabetes is paramount in developing effective public health and treatment strategies. The potential societal benefits are huge.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-gait-parameters-assessed-through-accelerometry-in-rheumatic-and-musculoskeletal-diseases-rmds

Analysis of Gait Parameters assessed through Accelerometry in Rheumatic and Musculoskeletal Diseases (RMDs)

Last updated:
ID:
47003
Start date:
13 February 2020
Project status:
Closed
Principal investigator:
Dr Mark Lunt
Lead institution:
University of Manchester, Great Britain

Painful rheumatic and musculoskeletal disease (RMD) conditions can impact the mobility and willingness to be active in patients. Furthermore, mobility is can be used as an indicator for fluctuations in disease severity. Passive recordings of patients’ symptoms can be used by clinicians to obtain a more objective and comprehensive image of disease progression and intervention effectiveness then only asking the patients about their pain in the last two months. Episodes of flare-ups, if not documented, might be forgotten by the next doctor visit or downplayed. Furthermore, activity trackers, such as fit-bit, are ever more frequently used in everyday life and patients receive methods for self-tracking their conditions positively. Therefore, developing Apps for smart-watches, which can then be recommended by clinicians, promises to become a helpful and cheap diagnostic tool.

We aim to identify walking behaviour and gait parameters that can be identified by such an App, using machine learning algorithms. Therefore we will take 2 years to compare parameters in patients with RMD conditions (such as osteoarthritis and rheumatoid arthritis) to healthy individuals, compare parameters between different RMDs and to observe the effect of covariates (such as obesity, medication history and self-reported physical activity) between individuals with the same RMD. The comparison between healthy and diseased individuals will help us to assess how reliably diseased individuals can be identified according to their gait pattern using wrist-worn accelerometers in real living conditions. Comparison between different RMDs can help further understand the different effect the diseases have on patient mobility. At last, conducting comparisons within the RMD groups allows for evaluating the effect of co-morbidities that affect an individual’s gait, such as obesity. If possible, indication of disease severity could also be used to identify the effect of disease severity on gait and walking behaviour.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-genetic-background-of-cholestatic-liver-diseases

Analysis of genetic background of cholestatic liver diseases

Last updated:
ID:
59089
Start date:
21 June 2021
Project status:
Current
Principal investigator:
Dr Marcin Krawczyk
Lead institution:
University of Saarland, Germany

The aim of our project is to investigate the genetic background of cholestatic liver diseases. This analysis focuses on identifying individuals within the UK Biobank with increased cholestasis markers in accordance with the guidelines of the European Association for the Study of the Liver. Previously, many studies showed that even small genetic variation may influence susceptibility and course of liver disease. Here, we would like to investigate whether determination of cholestatic disorders can be explained by inherited predisposition. These results will be coupled with the analyses of large cohorts of patients that we have been taking care for in our centres. We foresee that these approach may give rise to new diagnostic and therapeutic methods in patients with hepatobiliary diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-genetic-clinical-and-environmental-factors-from-the-uk-biobank-to-develop-new-approaches-in-the-prevention-and-treatment-of-pain-and-pain-related-diseases

Analysis of genetic, clinical and environmental factors from the UK Biobank to develop new approaches in the prevention and treatment of pain and pain-related diseases.

Last updated:
ID:
93305
Start date:
26 January 2023
Project status:
Current
Principal investigator:
Chanchal Kumar
Lead institution:
Grünenthal GmbH, Germany

Nearly one-fifth of adults worldwide, or about 1.5 billion people, suffer from chronic pain (CP), which in turn is frequently associated with several comorbidities and a decrease in quality of life. Although there are many pain relieving drugs available, there is still a high need of anti-pain medication for a number of painful diseases. Side effects and risk of drug addiction are an issue with many available products. Grünenthal has a vision to create a world free of pain by developing novel, safe and differentiated treatments for patients suffering from pain
It is very important for our research to have access to a combination of human genetic data, detailed patient information about pain and other relevant environmental data. This is difficult to acquire, as most available public health and genetic data about pain is from family illnesses or from animal studies, which is often not comparable with the condition of most patients. With the UK Biobank’s data on genetics, patient health records including pain phenotypes, biomarkers and environmental data it fills this gap and gives us the opportunity to explore new disease and treatment hypotheses for pain and related diseases.
To achieve this goal we want to explore genetic variation in the UK Biobank participants and the effect of those variations on pain and painful diseases to find genes that can be candidates for new pain treatments. This would also improve our understanding of pain diseases mechanisms in general. We plan the project to last at least 36 month, as disease progression over time it important for chronic pain diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-genetic-environmental-lifestyle-and-self-physical-health-risk-factors-for-ocular-complications-in-high-myopia-patients

Analysis of genetic, environmental, lifestyle and self-physical health risk factors for ocular complications in high myopia patients

Last updated:
ID:
105765
Start date:
3 July 2023
Project status:
Current
Principal investigator:
Dr Yanze Yu
Lead institution:
Fudan University, China

The aim of our study is to investigate the genetic, environmental, lifestyle, and self-physical health factors associated with ocular disorders in individuals with high myopia and to develop a predictive model to assess the risk of ocular complications in such patients. The study is motivated by the lack of understanding of the underlying risk factors leading to complications associated with high myopia. To address this gap in knowledge, the study will leverage the extensive genetic and health data available in the UK Biobank to uncover new insights into the risk factors associated with high myopia complications. The research methodology involves several analytical approaches, including genetic analysis, logistic regression, multivariate analysis, and machine learning. The project is expected to last for three years.
The findings of this research could have a significant impact on public health by improving screening and management of high myopia, potentially reducing the incidence of vision loss. Moreover, the development of predictive models could enhance clinical decision-making and improve patient outcomes. Overall, this study has the potential to advance our understanding of high myopia and its complications, leading to better prevention and treatment options for this prevalent and potentially serious condition.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-genetic-underpinnings-of-brain-network-features-in-health-and-in-psychotic-disorders

Analysis of genetic underpinnings of brain-network features in health and in psychotic disorders

Last updated:
ID:
56454
Start date:
16 February 2021
Project status:
Current
Principal investigator:
Dr Mikail Rubinov
Lead institution:
Vanderbilt University, United States of America

Complicated brain activity patterns allow us to perceive and respond to our environment in a uniquely human way. Poorly understood interactions between brain regions help to support the development, organization and function of these activity patterns. The organization of these interactions can differ significantly between healthy people, and people with neurological and psychiatric disorders, including psychotic disorders such as schizophrenia. These differences make it probable that abnormalities of such interactions contribute, at least in part, to the pathology and symptoms of brain disorders.

In turn, it is likely that specific interactions between brain regions are determined, in large part, by specific interactions between sets of genes. For example, recent studies have shown strong relationships between patterns of gene expression and brain activity. Our project will develop new computational methods, and will apply these methods to study detailed relationships between genes and brain activity. The project will investigate these relationships in healthy brains, and will use acquired knowledge of these relationships to better understand the organization of brain activity in psychiatric, including psychotic, disorders. Such improved understanding will ultimately help to gain insights into the genetic basis of healthy and diseased brain activity and function, and will, in this way, help make the diagnosis of psychiatric disorders more valid and objective.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-genotype-and-phenotype-data-from-the-uk-biobank-to-characterize-genes-and-pathways-impacting-human-disease-predict-efficacy-and-potential-adverse-outcomes-of-ionis-drug-targets

Analysis of genotype and phenotype data from the UK Biobank to characterize genes and pathways impacting human disease, predict efficacy, and potential adverse outcomes of Ionis drug targets.

Last updated:
ID:
94121
Start date:
6 December 2022
Project status:
Current
Principal investigator:
Dr Kavita Praveen
Lead institution:
Ionis Pharmaceuticals, Inc, United States of America

Drug targets with evidence of efficacy in humans, such as obtained from analyzing human genetic data, have a significantly higher likelihood of becoming approved, successful treatments. Hence it is now a necessity that pharmaceutical companies incorporate human genetics evidence when designing new therapeutics. We are applying to access the extensive health and medical data and genetic data collected by the UK Biobank in order to integrate human genetic findings into the drug discovery and development pipeline at Ionis. We will analyze human genetic data to identify novel drug targets, prioritize Ionis targets in development with supporting human genetic evidence or by discovering potential adverse effects, and to identify additional diseases that could benefit from existing Ionis drugs. Our analyses will include diseases in the cardiometabolic, renal, neuroscience, ophthalmology and immunology fields. Leveraging human genetics to identify targetable genes/pathways implicated in genetically complex, common diseases will help a much greater number of patients benefit from the therapeutic technologies that Ionis has successfully used for treatment of rare genetic disorders. We hope that incorporating human genetic evidence in our therapeutic pipeline will help streamline and accelerate the development of successful and safe therapies at Ionis, thus benefiting more patients, and faster.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-human-daily-activity-cognitive-function-and-well-being

Analysis of human daily activity, cognitive function, and well-being.

Last updated:
ID:
46409
Start date:
29 May 2019
Project status:
Closed
Principal investigator:
Dr Masato S. Abe
Lead institution:
RIKEN, Japan

Human behaviors emerge from interactions between brains and the environment. Thus, human daily activity can be associated with cognitive brain functions and subjective well-being. However, the relationship between them remains unknown. In this project, we will analyze accelerometer data, and reveal how the characteristics of activity data in daily life reflect cognitive functions and subjective well-being. To analyze time-series accelerometer data we will use statistical mechanics and machine learning method. The former method can give us a distribution of consecutive activity or inactivity in time-series. Previous studies revealed that the distribution followed a power law distribution and the exponents were different between healthy subjects and those with mental illness (Nakamura et al. 2007, Physical Review Letter; Sano et al. 2011, Plos one). The latter method is useful for predicting time-series data and outcomes. By predicting the future values from past data, we can characterize the predictability of human daily life and how complex the daily activity is. Finally, we will conduct statistical analysis for revealing the relationship between the parameters extracted from time-series data and other characteristics of subjects. The project duration will be 18 months. Quantifying how accelerometer data correlates with the subject’s daily life patterns and well-being can provide novel insights into human behavior research field and health care. The possible application includes finding biomarkers of human well-being only from the time-series of accelerometer data.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-large-scale-genotype-and-phenotype-data-to-facilitate-the-discovery-and-development-of-base-editing-therapeutics

Analysis of large-scale genotype and phenotype data to facilitate the discovery and development of base editing therapeutics

Last updated:
ID:
84811
Start date:
30 March 2022
Project status:
Current
Principal investigator:
Dr Margaret Parker
Lead institution:
Beam Therapeutics, United States of America

By far, the largest class of disease-causing mutations involves changes to single DNA base pairs. Most diseases caused by single base pair changes cannot currently be treated with traditional therapeutics, such as small molecule drugs. Thus, developing therapeutics to directly correct disease-causing mutations is of great interest for the treatment of genetic diseases. Recently developed gene-editing technologies, such as base editing, can directly create base substitutions in cellular DNA and therefore safely and efficiently reverse the effect of pathogenic mutations. However, because there are thousands of diseases caused by single base pair changes, it is important to be able to prioritize specific diseases that have the greatest unmet need and which are most likely to be successfully targeted by gene editing approaches. This research aims to use the UK Biobank to inform these decisions. We expect results of this application to include: 1) the discovery of new mutations and genes associated with disease risk; 2) improved understanding of penetrance and clinical presentation of carriers with known disease-causing mutations; and 3) improved understanding of the safety of gene editing therapeutics. This research will advance the development of a new class of therapeutics (base editors), ultimately benefiting patients and contributing to our understanding of the consequences of genetic variation on disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-lifestyle-genetic-and-metabolic-factors-in-the-uk-biobank-for-understanding-and-predicting-chronic-diseases

Analysis of Lifestyle, Genetic, and Metabolic Factors in the UK Biobank for Understanding and Predicting Chronic Diseases

Last updated:
ID:
635828
Start date:
21 July 2025
Project status:
Current
Principal investigator:
Miss Xingjun Cai
Lead institution:
Hainan Medical University, China

(I) Scientific Problems
Chronic diseases like cardiovascular disease and diabetes are rising, but their complex pathogenesis involves lifestyle, genetic susceptibility, and metabolic disorders. The interaction mechanisms among these factors remain unclear. Lifestyle factors (e.g., diet, exercise, smoking) impact individuals differently, complicating personalized prevention strategies.
(II) Objectives
Investigate the associations and interaction mechanisms between lifestyle, genetic factors, and metabolic disorders in the risk of common chronic diseases (e.g., cardiovascular disease, diabetes, chronic respiratory diseases).
Develop a risk assessment model for chronic diseases based on multi-omics data (e.g., genetic and metabolic data) and lifestyle information to enable early risk prediction.
Provide a scientific basis for personalized prevention and intervention strategies, including tailored lifestyle improvements and treatments.
(III) Scientific Basis
Epidemiological studies show that unhealthy lifestyles (e.g., high-sugar/high-fat diets, inactivity, smoking) are key risk factors for chronic diseases, but their synergistic effects and disease-specific contributions are unclear. Genetic factors significantly influence chronic disease development, with GWAS identifying many associated loci. However, how genetic variations interact with environmental factors to affect disease susceptibility requires further study. Metabolic disorders (e.g., abnormal glucose, lipids, blood pressure) are crucial for chronic disease pathophysiology. Blood biomarkers reflect metabolic states and may indicate early disease stages before clinical symptoms, aiding diagnosis and monitoring. Improved sensitivity and specificity of biomarkers and optimized biomarker combinations or prediction models are needed for better early detection and monitoring.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-long-term-health-risks-identifying-key-factors-for-chronic-diseases

Analysis of Long-term Health Risks: Identifying Key Factors for Chronic Diseases

Last updated:
ID:
349386
Start date:
11 December 2024
Project status:
Current
Principal investigator:
Professor Chen Dong
Lead institution:
Soochow University, China

Chronic diseases, such as cardiovascular disease, diabetes, and liver disease, are major health concerns that affect millions of people worldwide. Despite our current knowledge, there are still many unanswered questions about what factors contribute to the development of these diseases.

The aim of this research project is to investigate the relationships between various risk factors and the likelihood of developing chronic diseases. We will use the data from the UK Biobank, a large-scale study that has collected detailed information, including genetic, lifestyle, and biomedical data, as well as metabolite profiles, from a large number of participants. By identifying and understanding the complex interplay between various risk factors, we can gain deeper insights into the underlying biological mechanisms that contribute to chronic disease development. This knowledge can then be leveraged to develop more effective and personalized prevention strategies, targeting individuals or groups with specific risk profiles.

This research is expected to take approximately 3 years to complete.

The potential public health impact of this research is significant. By enabling early identification of individuals at high risk for chronic diseases, our findings could inform the development of targeted prevention programs, early intervention strategies, and personalized treatment approaches. Ultimately, this research has the potential to reduce the burden of chronic diseases on individuals, healthcare systems, and society as a whole, leading to improved health outcomes and quality of life for millions of people.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-mitochondrial-heteroplasmy-and-its-association-to-age-and-health-conditions

Analysis of mitochondrial heteroplasmy and its association to age and health conditions

Last updated:
ID:
59252
Start date:
8 February 2021
Project status:
Current
Principal investigator:
Professor Sangwoo Kim
Lead institution:
Yonsei University, Korea (South)

During an entire lifetime, somatic mutations occur in DNA, which are the causes of many disease including cancer, complex diseases and probably ageing. While somatic mutations in genomic DNA have been widely analyzed from whole-genome or whole-exome sequencing, those that occur in mitochondrial genome have been rarely assessed due to the technical difficulties. As mitochondrial genome encodes many protein coding genes for energy metabolism and electron transport chain, mutations in mitochondrial genome and its resulting breakdown of genes may be associated or causative of related diseases. In this project we will develop a robust algorithm to assess somatic mutations in mitochondrial genome and use it for its clinical value for detecting, diagnosing or preventing human diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-phenotypic-and-environmental-factors-associated-with-identified-genetic-variants-in-skin-cancer

Analysis of phenotypic and environmental factors associated with identified genetic variants in skin cancer.

Last updated:
ID:
92702
Start date:
29 March 2023
Project status:
Current
Principal investigator:
Dr Kavita Sarin
Lead institution:
Stanford University, United States of America

Skin cancer is the most common human malignancy with rising incidence rates. We are focused on understanding the genetics behind skin cancer (both melanoma and non-melanoma) to develop new targeted therapies. We have made significant progress in identifying specific genetic variants that play a role in squamous cell carcinoma, basal cell carcinoma, melanoma, and other cancer-associated skin diseases. The aims of this project are to use the UK Biobank to 1) validate the impact of genetic variants associated with skin cancer, 2) explore how cancer-related variants effect associated phenotypes such as frequency, coloration, and size of cutaneous lesions, 3) understand how environmental exposures may modulate the effect of genetic variants on skin cancer, and 4) identify if identified genetic variants increase the risk for other malignancies. We will study variants from different skin diseases separately using statistical tests to determine their association with phenotypic, environmental, and biomarkers measurements. Reviewing evidence from the UKBiobank will increase our confidence regarding which variants may serve as potential therapeutic targets to improve skin cancer outcomes.

Results from our analyses will guide subsequent functional experiments aimed at understanding which downstream signaling pathways are affected, and how these variants synergize with known oncogenic pathways (i.e., p53, ATK, MAPK pathways). Because this project involves multiple studies focused on different skin diseases, we request data access for the maximum project duration of 36 months.

The public health impact of this project is to extend our understanding of skin cancer genetics and improve our annotation of specific genetic mutations enriched in skin cancer. Furthermore, the proposed project will enable us to identify druggable targets, thereby facilitating the development of new therapeutics to improve skin cancer outcomes.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-plasma-proteomics-data-for-age-related-macular-degeneration-amd-donors

Analysis of plasma proteomics data for Age-Related Macular Degeneration (AMD) donors

Last updated:
ID:
637144
Start date:
17 October 2025
Project status:
Current
Principal investigator:
Dr Aurelian Radu
Lead institution:
Icahn School of Medicine at Mount Sinai, United States of America

Objectives. The goal is to analyze the proteomics data generated by the O-link strategy (full set of plasma proteins) from donors diagnosed with AMD in comparison with non-AMD donors.
Rationale. A plausible hypothesis is that systemic processes contribute to the local degenerative changes that affect the retina in AMD. The systemic processes could be reflected by changes in the abundance of some plasma proteins. The differentially-abundant plasma proteins could provide information about the cellular and molecular mechanisms that contribute to AMD, which are largely unknown.
Research questions. The questions are whether some plasma proteins have significantly different abundance in AMD, and if they can be related to AMD mechanisms.
Approach. We propose to analyze the O-link data available in the UK Biobank (Record Table 1072), and more specifically the subset derived from AMD donors (>300 cases), for all the proteins detectable by the O-link technology. The set will be compared to 300-500 cases of donors that did not have AMD. The controls will be selected to have a similar distribution for age, gender and ethnicity. Additionally, information regarding AMD and known risk factors will potentiate the analysis (eye exam, fundus and OCR images, presence of genetic risk alleles for AMD, smoking status, BMI, hypertension, diabetes).
The differentially abundant proteins will be analyzed for pathways that could be relevant for AMD. If the results are promising, the time frame of the analysis will be extended, to take advantage of the O-link data that continue to accumulate. The project requires only de-identified data.
Potential impact. Differences in the abundance of some plasma proteins could shed light on the mechanisms that lead to AMD and its progression. This could in turn suggest interventions to inhibit AMD progression. Some of these differences could have a causal role in AMD and therefore correction of the differences could have therapeutic potential.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-population-admixture-and-its-implications-for-studies-of-dysmenorrhoea

Analysis of Population Admixture and Its Implications for Studies of Dysmenorrhoea

Last updated:
ID:
50708
Start date:
30 October 2019
Project status:
Closed
Principal investigator:
Dr Zhiqiang Li
Lead institution:
Affiliated Hospital of Qingdao University, China

Dysmenorrhoea is a medical condition characterized by menstrual pain, and Its aetiology and pathophysiology remain largely unknown. This project is intended to identify genetic factors for dysmenorrhoea by leveraging multi-ancestry genome-wide data. We will use data from both of UK Biobank (European) and our own study (Chinese). The trans-ethnic genetic analysis from different populations is proven to increase power for detecting genetic factors. The duration of this project is about six months. This project may provide new insights into the understanding of dysmenorrhoea and facilitate management of the condition.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-potential-pleiotropic-effects-of-specific-target-genes-of-interest-through-a-comprehensive-phewas-analysis-across-a-large-population

Analysis of potential pleiotropic effects of specific target genes of interest, through a comprehensive PheWAS analysis across a large population.

Last updated:
ID:
101646
Start date:
6 July 2023
Project status:
Current
Principal investigator:
Dr David Torrents
Lead institution:
Barcelona Supercomputing Center, Spain

Complex diseases, which are caused by the interaction of multiple genes, such as asthma, type 2 diabetes, or some skin disorders have been broadly studied at the genomic level during the last decades. As a result, thousands of genomic variants have been found associated with these diseases, thus enhancing their detection, prevention, and treatment. However, there are still many genomic studies that can be done to complement and improve the genetic knowledge of complex diseases. In this direction, we have developed new methods which can enhance the study of the effect of genomic variants in multiple traits or diseases, which is also named pleiotropy.

The common way to study pleiotropy is to test if a variant is simultaneously associated with two or more related traits. Therefore, to find pleiotropic variants it is necessary to perform an association test with diverse phenotypes. To do this type of analysis, we have currently developed and published a method, which facilitates the inclusion and, therefore, the genotype-phenotype test of more good quality genomic variants. Therefore, although some large-scale initiatives have already globally inspected pleiotropy based on previously published association studies results, there are still some variants which have been completely disregarded or even excluded from the analyses.

To find pleiotropic variants associated with some skin disorders, we will focus on a reduced set of variants with a certain known relation with genes linked with the disease. Then, we will use the UK Biobank cohort to test the association between these variants and diverse traits and complex diseases. As a result, we will obtain a list of variants associated with multiple traits or diseases.

Given the complexity of the project, its estimated duration is of 3 years. The outcomes of these analyses are expected to provide a better understanding of the pathophysiology of complex diseases, to result in high impact scientific publications, and to ultimately help diagnose and treat complex diseases in a personalised manner. Particularly, being in a collaboration with a pharmaceutical company, Almirall, these outcomes will be evaluated for the generation of drug discovery programs and, therefore, to produce new and/or more effective therapies for skin diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-rare-variants-in-immune-related-genes-associated-with-clinical-outcomes-in-neurodegeneration-and-cancer

Analysis of rare variants in immune-related genes associated with clinical outcomes in neurodegeneration and cancer.

Last updated:
ID:
52361
Start date:
25 October 2019
Project status:
Closed
Principal investigator:
Dr Shameek Biswas
Lead institution:
Celgene Corp., United States of America

We intend to study the role of immunological variation existing in the human population in modulating progression of diseases that have a significant immune component (cancer and neurodegeneration). There have been a number of large genetic studies that have characterized the impact of immune system on disease risk and our focus would be to extend this to disease progression in cancer and neurodegeneration. Genetic variants that affect the activity of molecule within cells can act as surrogates to model the potential effects of therapeutic intervention in a randomized clinical trial. With the integration of longitudinal assessments, we will use genetics to infer the potential therapeutic impact of targeting certain molecules in silico.
At a global level, we aim to answer this question by testing whether genetic risk from autoimmune disease associates with cancer survival. At a gene level, we will ask the same question for a subset of rare, high impact functional immune genetic variants. For neurodegeneration, we intend to prioritize genes that are enriched for expression in astrocytes and microglia, two cell types that appear important for Alzheimer’s disease risk.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-risk-factors-and-long-term-prognosis-of-central-nervous-system-inflammatory-diseases

Analysis of Risk Factors and Long-term Prognosis of Central Nervous System Inflammatory Diseases

Last updated:
ID:
477093
Start date:
14 March 2025
Project status:
Current
Principal investigator:
Professor Yu Rang Park
Lead institution:
Yonsei University, Korea (South)

Objective: To retrospectively analyze a cohort of patients with central nervous system (CNS) inflammation to determine the occurrence of neurological complications.

Primary Endpoint: The occurrence of comorbidities and the time to comorbidity onset during a follow-up period of at least 5 years.

Secondary Endpoint: Time from diagnosis of CNS inflammatory disease to death.

Data Source: UK Biobank data.

Definitions:

CNS Inflammation: Approximately 40,000 adult patients diagnosed with CNS inflammatory diseases (based on ICD codes) and hospitalized between 2008 and 2017.
Control Group: Patients diagnosed with headaches between 2008 and 2017.
Comorbidities: Neurological and psychiatric conditions such as Amyotrophic Lateral Sclerosis, Brain Tumor, Hemorrhagic Stroke, Ischemic Stroke, Alzheimer’s Disease, Dementia, Parkinson’s Disease, Multiple Sclerosis, Major Depressive Disorder, Anxiety Disorder, Epilepsy, Neuromyelitis Optica, among others.
Analysis:

Survival analysis adjusted for age and sex.
Subgroup Analysis: Stratification by pathogen group where possible.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-risk-factors-and-prognostic-factors-related-to-prostatic-diseases-prostatitis-benign-prostate-hyperplasia-and-prostate-cancer

Analysis of risk factors and prognostic factors related to prostatic diseases (prostatitis, benign prostate hyperplasia and prostate cancer)

Last updated:
ID:
96548
Start date:
9 February 2023
Project status:
Current
Principal investigator:
Professor Xian-Tao Zeng
Lead institution:
Wuhan University, China

Aims:
1) To identify potential risk factors within UK Biobank to determine whether they are causally associated with development of prostatitis, benign prostatic hyperplasia and prostate cancer.
2) To identify potential prognostic factors within UK Biobank to determine whether they are associated with short-term and long-term outcome of prostatitis, benign prostatic hyperplasia and prostate cancer.
3) To calculate polygenic risk scores for exploring its effect and interaction with non-genetic factors, and measuring potential causality.
4) To develop and validate risk prediction models for prostatitis, benign prostatic hyperplasia and prostate cancer.
5) To explore trajectory of risk and prognostic factors and their causal associations with development and prognosis of prostatitis, benign prostatic hyperplasia and prostate cancer.

Scientific rationale:
Populations are growing older in countries and population ageing has been a global phenomenon. Prostate disease incidence directly correlates with age. National disease burden of the three prostatic diseases will continue to increase. In our previous studies, we had found that gut microbiota, cardiovascular disease, oral microbiome, periodontal disease, body mass index, gene polymorphisms were associated with prostate disease. However, there is still a lack of sufficient and robust evidence on broadly estimating risk and prognosis of the prostate diseases and the trajectory changes of these factors and disease in a population-based prospective cohort. Moreover, the evidence of impacts of the factors on long-term outcome of the prostatic diseases are also insufficient.

Project duration
This project is expected to last for 36 months.

Public health impact
This work will be an advancement in our understanding of development and prognosis of prostatitis, benign prostatic hyperplasia and prostate cancer. Knowledge of identified links can enable the public, policymakers and epidemiology scientists to make preventative strategies for the three prostatic diseases and can provide scientific evidence for early intervention and better management.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-risk-factors-for-hematopoietic-and-immune-system-aging-and-their-association-with-immune-diseases-and-cancer

Analysis of risk factors for hematopoietic and immune system aging and their association with immune diseases and cancer

Last updated:
ID:
106397
Start date:
23 June 2023
Project status:
Current
Principal investigator:
Dr Qiang Tong
Lead institution:
Shanghai Sixth People's Hospital, China

The purpose of this study is to understand how different parts of the body age and affect diseases. By learning about the causes of aging, we hope to find ways to prevent or treat diseases. This study will take around 12 months to 3 year. We will look at how genes, lifestyle, and diet affect aging. By making positive changes in these areas, we can delay aging and prevent disease, and improve health outcomes.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-shared-genetic-determinants-across-major-complex-human-disorders

Analysis of shared genetic determinants across major complex human disorders

Last updated:
ID:
36647
Start date:
19 May 2020
Project status:
Current
Principal investigator:
Professor Andrey Rzhetsky
Lead institution:
University of Chicago, United States of America

We are aiming to design a method where all diseases can genetically mapped in a single analysis, as opposed to disease-by-disease mapping. We then perform this mapping and use it to predict patient’s future health from her genetics.

Public health impact of the study

Our study (hopefully) will pave way to discovering how the same genetic variant affects multiple diseases. This, in turn, should increase the quality of disease forecasting from individual-specific genetic data. Our hope is that high-quality forecast of likely future pathologies facing particular individual would allow to design patient-specific preventive measures.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-sleep-phenotypes-genotypes-and-associated-conditions

Analysis of sleep phenotypes, genotypes and associated conditions

Last updated:
ID:
19705
Start date:
1 March 2017
Project status:
Closed
Principal investigator:
Dr Guy Leschziner
Lead institution:
Guy's and St Thomas' NHS Foundation Trust, Great Britain

Insomnia is increasingly understood to be associated with a variety of other sleep conditions, as well as cardiovascular disease, dementia and psychiatric illness. We aim to better understand the nature and direction of these associations, and to identify genetic contributions to insomnia and other sleep traits. We also intend to see if there are subgroups of individuals with different types of insomnia, based upon phenotype and genotype. An understanding of the relationship between sleep disorders and disease of major public health interest will hopefully focus attention on a potentially modifiable set of risk factors for ischaemic heart disease, stroke, hypertension, dementia and psychiatric disease. Identifying genetic polymorphisms underlying insomnia may permit the development of new therapeutic targets based upon newly discovered biochemical or genetic pathways. We aim to analyse genetic and neuroimaging data according to the presence or absence of insomnia and associated sleep traits, to identify common genetic variants that increase susceptibility to insomnia. There is evidence that insomnia can be subdivided into different subgroups based upon psychological trait data and psychiatric co-morbidity, and we aim to undertake cluster analysis on the whole cohort to see if particular genetic variants are correlated with particular psychological/psychiatric trait data. We also aim to investigate the correlations with neuroimaging data where available. We would like to include the whole cohort.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-the-association-and-causality-of-glaucoma-and-risk-factors-exposure-with-combined-of-analyses-of-genome-and-image-study

Analysis of the association and causality of glaucoma and risk factors (exposure) with combined of analyses of genome and image study

Last updated:
ID:
96390
Start date:
23 January 2023
Project status:
Current
Principal investigator:
Dr Jehyun Seo
Lead institution:
Veterans Health Service Medical Center, Korea (South)

The aim of this project is to investigate the association and causal relationship between glaucoma and previously-suggested risk factors using a combination of genomic and image analysis.
The scientific rationale for this study is that prospective cohort or randomization control trial data are required to verify the causation of glaucoma risk factor, however, these data are difficult to obtain. Mendelian Randomization (MR) analysis, a recently developed statistical model, can be used to establish causality for cohorts containing genetic data. In this study, targeted risk factors for glaucoma (intarocular pressure, diabetes, systemic hypertension or hypotension, cornea thickness, corneal hysterosis, myopia, body mass index, estimated cerebrospinal fluid pressure, sleep apnea, cardiovascular data, imaging data, lab data) will be assessed by MR analysis and genome-wide association study. Furthermore, using UK Biobank data, a genome-based glaucoma prediction model analysis will be constructed, as will a study to determine the reason of missing heritability and rare variant analysis The researchers estimate the project will take three years. Risk factors with confirmed causality beyond the level of association are chosen for this project, and it is hoped that they will be used in public health effect for the prevention of glaucoma, which is a leading cause of irreversible blindness worldwide. It is anticipated that it will be possible to give data that forms the basis for precision medicine, which delivers genome-based illness information, by investigating the association between these risk factors and glaucoma.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-the-association-and-causality-of-sleep-patterns-psychological-states-and-cognitive-function-with-combined-of-analyses-of-genome-and-image-study-a-prospective-cohort-study-of-uk-biobank

Analysis of the association and causality of Sleep Patterns, psychological states and Cognitive Function with combined of analyses of genome and image study: a prospective cohort study of UK biobank

Last updated:
ID:
544746
Start date:
24 March 2025
Project status:
Current
Principal investigator:
Professor Xia Li
Lead institution:
Shanghai Jiao Tong University School of Medicine., China

According to the latest data and research, the prevalence of dementia is steadily rising worldwide. It is projected that by 2030, the number of individuals affected by dementia will surge to 78 million. Dementia is not only a significant health concern but also imposes a substantial economic burden globally. In 2019, the economic losses attributed to dementia amounted to 1.3trillion. This situation presents considerable challenges for individuals, families, and society at large. Therefore, it is imperative to implement effective preventive measures and the standards of diagnosis and treatment for dementia. In preliminary studies, we identified sleep disturbances as risk factors for dementia. However, the trajectory of sleep disturbances preceding dementia, as well as the mechanis.
This study will analyze an elderly sleep cohort to: 1) explore sleep patterns among older adults across different age groups, genders, and APOE gene carriers; 2) investigate the role of APOE gene variations in the imaging of sleep disorders and their psychological and cognitive implications; 3) delineate the brain network trajectories associated with psychological and cognitive impairments resulting from sleep disorders; and 4) establish a multimodal predictive model for psychological and cognitive impairments based on sleep disturbances. This comprehensive approach allowed us to delve deeper into the relationship among sleep disturbances, mental disorders and cognitive impairment in the aging.
We expect that this project will take 36 months to complete. Investigating the relationship between sleep and psychological-cognitive function, along with the underlying molecular and neurofunctional mechanisms, is crucial for advancing research on the pathogenesis of cognitive impairments. This understanding holds significant practical value for preclinical interventions targeting cognitive disorders and offers considerable societal benefits for the harmonious development of an aging population.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-the-association-between-genetic-variation-linked-to-neurological-disorders-and-infectious-disease-risk

Analysis of the association between genetic variation linked to neurological disorders and infectious disease risk

Last updated:
ID:
654653
Start date:
30 June 2025
Project status:
Current
Principal investigator:
Mr Oiher Serrano Asensio
Lead institution:
University College London, Great Britain

We aim to define new pathogenic mutations that cause Parkinson’s disease (PD) and other neurological disorders and to investigate the overlap between neurological disorders and risk / protection for infectious diseases. We will evaluate genetic, biomarker and clinical data regarding infectious disease history to investigate the link between rare variants in lysosomal and immune-related genes associated with neurological movement disorders, such as LRRK2 or GBA1 in Parkinson’s disease. The rationale for this research is that there are mutations that are protective against infectious diseases such as tuberculosis and leprosy, which may become common due to selective pressure to survive and reproduce in early life, could affect risk of developing neurological disorders later in life. Thus, genetic pleiotropy may apply to both common and rare variants. We propose the following research questions:
-What is the UK Biobank population prevalence and penetrance of known and novel genetic and biomarker (SOMASCAN / OLINK) risk factors for neurological diseases including Parkinson’s disease, PSP, and dementia – we will analyse this data in relation to data generated in the global Parkinson’s genetics programme and 100K genomes programme.
-Do carriers of these variants have a clinical history of infectious disease, including tuberculosis and mycobacterial infection (i.e. leprosy) in comparison to non-risk allele carriers (controls)
-What is the ancestry distribution of these carriers, and which infectious disease types and rare variants are more prevalent in each ancestry
-Is there an association between genetic ancestry, infectious disease risk, and lysosomal/immune gene rare variant type. Does this correlate with differences in neurological disease penetrance and outcomes (e.g. severity, duration)


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-the-association-between-hearing-loss-and-tinnitus-and-metabolic-diseases-metabolite-correlations

Analysis of the association between hearing loss and tinnitus and metabolic diseases & Metabolite Correlations

Last updated:
ID:
146430
Start date:
28 February 2024
Project status:
Current
Principal investigator:
Professor Incheol Seo
Lead institution:
Kyungpook National University, Korea (South)

We aim to better understand the relationships between tinnitus (ringing in the ears), hearing loss and metabolic diseases. By analyzing extensive data from the UK Biobank, we will investigate if people with metabolic conditions are more likely to experience hearing loss and tinnitus and if specific metabolic pathways are involved. This study could identify substances in the body (metabolites) that could be related to hearing loss and tinnitus. Our findings might lead to earlier detection and new treatments for hearing loss and tinnitus. The project will last for 36 months, during which we will analyze data and share our discoveries to benefit public health.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-the-composition-of-blood-immune-cells-in-blood-clot-of-large-artery-atherosclerosis

Analysis of the composition of blood immune cells in blood clot of large artery atherosclerosis

Last updated:
ID:
177007
Start date:
26 November 2024
Project status:
Current
Principal investigator:
Dr Bo Kyung Yoon
Lead institution:
Yonsei University, Korea (South)

Aims:
The aim of this research is to explore the blood profile, including metabolite levels, of ischemic stroke patients with different underlying causes. Specifically, we will investigate how the transcriptomic profiles of macrophages in blood clots vary depending on the etiology of the stroke. We also aim to understand how blood cholesterol levels influence macrophage activity in these patients.

Scientific Rationale:
Ischemic stroke, caused by a blockage in the blood vessels supplying the brain, can result from various underlying conditions. Recent findings suggest that the immune cell composition within blood clots differs based on the cause of the stroke. Our preliminary research has shown distinct gene expression profiles of macrophages in clots from different etiologies. Macrophages play a crucial role in stroke pathology, and their activity is known to be influenced by blood cholesterol levels, as seen in foamy macrophages in atherosclerosis plaques. By investigating the blood profiles of stroke patients, we hope to uncover how cholesterol and other metabolites affect macrophage behavior and contribute to stroke outcomes.

Project Duration:
This project is expected to take three years to complete, including data analysis, interpretation, and dissemination of findings.

Public Health Impact:
Understanding the relationship between blood metabolites and macrophage activity in ischemic stroke patients could lead to significant advancements in stroke diagnosis, treatment, and prevention. By identifying specific metabolic profiles associated with different stroke etiologies, we can develop targeted therapies to modulate macrophage activity, potentially improving recovery and reducing the risk of recurrent strokes. This research has the potential to enhance personalized medicine approaches for stroke patients, ultimately improving public health outcomes and reducing the burden of this debilitating condition.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-the-dynamic-characteristics-of-aging-related-serum-uric-acid-in-nervous-and-digestive-system-disorders-with-machine-learning-methods

Analysis of the Dynamic Characteristics of Aging-related Serum Uric Acid in Nervous and Digestive System Disorders with Machine Learning Methods

Last updated:
ID:
90369
Start date:
8 September 2022
Project status:
Current
Principal investigator:
Professor Xiaoshan Zhao
Lead institution:
Southern Medical University, China

How to reduce chronic diseases in the aging process and promote “healthy aging” are important social public health issues. The nervous and digestive disorders are closely related to the aging process. However, the mechanism is still unknown. Previous studies have reported that the human nervous and digestive systems are interdependent. Our research team have found that uric acid is influenced by many factors in the aging process of healthy people. In this three-year project, we aim to conduct the following research: 1) Characterization of the dynamic changes of serum uric acid in health, the nervous and digestive disorders during aging process in a large cohort with additional validation datasets. 2) Demonstration of causal relationship between serum uric acid and the nervous and digestive disorders outcomes based on an integrated analysis of lifestyle, environmental exposures, genotypes, and imaging data using machine learning methods. The public health impact of this three-year study could help improve early diagnosis and treatment decisions for the nervous and digestive disorders associated with elevated serum uric acid, especially those chronic functional diseases that still lack clear objective criteria.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-the-effects-of-genetically-predicted-biological-pathways-on-right-heart-imaging-traits-in-relation-to-sex-and-the-interdependence-with-the-left-heart

Analysis of the effects of genetically predicted biological pathways on right heart imaging traits in relation to sex and the interdependence with the left heart.

Last updated:
ID:
95677
Start date:
11 May 2023
Project status:
Current
Principal investigator:
Dr Lars Harbaum
Lead institution:
University Medical Center Hamburg-Eppendorf, Germany

The right side of the heart pumps the blood through the lungs into the left heart. Many diseases of the lungs or the left side of the heart, many of which affect a large number of individuals, change the structure and function of the right heart. Measuring the right heart by imagine techniques such as cardiac magnetic resonance provides crucial information on patients’ current health status and risk for future events. This is mostly irrespective of the underlying disease. Our three-year project aims to understand the biology of the right heart. We will use genetic variables that reflect involvement of well-defined biological elements and carefully assess the relationship to the right heart measurements. We will separately investigate females and males to find gender-related differences. Understanding of the biology behind changes of right heart function and structure is the first step to develop drugs that specifically improve right heart function or blood-borne tests that directly inform on the status of the right heart.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-the-features-related-to-healthy-aging

Analysis of the features related to healthy aging

Last updated:
ID:
86776
Start date:
25 August 2022
Project status:
Current
Principal investigator:
Professor Tzu-Hao Chang
Lead institution:
Taipei Medical University, Taiwan, Province of China

The aim of this research is to establish the machine learning based predictive models to predict elderlies with subjective cognitive decline or mild cognitive impairment at high risk of future neurodegenerative progression through MRI and/or patient genotyping. Imaging features may serve as surrogates for certain outcome phenotypes. Genetic signatures can thus be linked to imaging features.
Specifically, the longitudinal MRI and genetic signatures of patients will be statistical analyzed to reveal the clinical risks of neurodegenerative effects in the individual level. Then, we propose to use the machine learning-based algorithm to model the longitudinal disease progression of the patients. Finally, the constructed machine learning-based predictive models can provide individualized diagnostic and therapeutic strategies in each patient. Combining genetic profiles and imaging features also has powerful synergistic potential in terms of risk stratification and precision medicine, proving opportunities for early, targeted intervention and prognostication.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-the-impact-of-different-blood-pressure-variability-on-imaging-hypoperfusion-in-elderly-hypertensive-population

Analysis of the Impact of Different Blood Pressure Variability on Imaging Hypoperfusion in Elderly Hypertensive Population

Last updated:
ID:
597548
Start date:
1 April 2025
Project status:
Current
Principal investigator:
Miss Junqi Yang
Lead institution:
The Eighth Affiliated Hospital of Sun Yat-sen University, China

I. Research Questions
1.To what degree does diverse blood pressure variability (BPV) impact imaging hypoperfusion in elderly hypertensives?
2. Which BPV types most closely correlate with hypoperfusion incidence/severity? Analyzing patterns from circadian rhythms, postural shifts, and medications will spotlight key risk factors.
3. Can BPV independently predict hypoperfusion post confounding factor adjustment?
II. Research Objectives
1.Comprehensively analyze BPV index distributions in elderly hypertensives. Assess mean, median, SD, and frequency of 24-hour BPV (SD, CV), nocturnal dip patterns, and visit-to-visit fluctuations. Identifying high-risk subgroups via these distributions enables proactive screening and early action.
2.Define the relationship between BPV levels and hypoperfusion presence/extent. MRI detects cerebral hypoperfusion via reduced CBF, CBV, T2-weighted or FLAIR images, diffusion-weighted imaging abnormalities. Multivariate regression and ROC curves will affirm this relationship’s clinical and prognostic value, steering personalized treatment strategies.
3.Construct a solid scientific basis for individualized elderly hypertensive care, monitoring and tracking. By clarifying BPV-hypoperfusion dynamics, advocate for personalized custom BP targets, drug regimens, and vigilant monitoring.
III. Scientific Rationale
Among the elderly, the surging prevalence of hypertension, compounded by intricate management demands and comorbidities like frailty, urgently necessitates a profound exploration of factors contributing to target organ damage. BPV’s link to cardiovascular outcomes is known, yet its role in cerebral perfusion needs exploration. New pathophysiological paths may emerge. The UK Biobank’s rich data provides an opportunity to bridge research gaps.Understanding BPV’s impact on hypoperfusion enables recalibrating antihypertensive targets, reducing morbidity and mortality and optimizing care for elderly hypertensives.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-the-impact-of-innate-immune-system-genetics-on-neurodegenerative-disease-and-cancer

Analysis of the impact of innate immune system genetics on neurodegenerative disease and cancer.

Last updated:
ID:
87390
Start date:
15 September 2022
Project status:
Current
Principal investigator:
Dr Gregory Minevich
Lead institution:
Alector LLC, United States of America

Aims:
We aim to assess the impact of genetic variants that control microglia function on brain health, thus identifying targets that may lead to therapies for neurodegenerative brain diseases.

Scientific rationale:
A growing body of human genetics research is revealing that many of the risk genes associated with neurodegenerative conditions are expressed in microglia, the immune cells of the brain. Microglia are critical for maintaining a healthy brain and for counteracting brain diseases. When microglia become dysfunctional due to normal aging or genetic factors, neurodegenerative diseases develop.

Expected duration:
The project is expected to take three years.

Public health impact:
We aim to identify/prioritize targets for new drug development, more accurately classify patients by combinations of genotype/phenotype for clinical trials, and develop readouts that we can use to assess how well our drugs may be restoring brain health.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-the-incidence-rate-risk-factors-related-surgical-strategies-postoperative-complications-and-prognosis-of-urogenital-system-diseases

Analysis of the incidence rate, risk factors, related surgical strategies, postoperative complications and prognosis of urogenital system diseases

Last updated:
ID:
529233
Start date:
18 March 2025
Project status:
Current
Principal investigator:
Dr Zhao Wang
Lead institution:
Xiangya Hospital of Central South University, China

This research project aims to investigate the incidence rate, risk factors, surgical strategies, postoperative complications and prognosis of some urogenital system diseases.By analyzing the data from the UK Biobank (a large biomedical database), we hope to reveal the impact of urogenital system diseases on public health.
A variety of genitourinary disorders are common in clinical practice, but there are no standardized criteria for their clinical diagnosis, classification and treatment, and there are inconsistencies in the relevant content of different textbooks or guidelines. In order to solve these problems, we will use the data from UKB to analyze some urinary system diseases (including benign prostatic hyperplasia, urinary calculi, urethral stricture, urinary incontinence, neurogenic bladder, ketamine-related cystitis and radiation cystitis, etc.), male and female reproductive system diseases (including genital malformations of both sexes, male infertility and female infertility, etc.), female pelvic organ prolapse-related diseases, surgical strategies (such as for patients with refractory OAB, whether to choose botulinum toxin injection, sacral nerve modulation or tibial nerve stimulation; comparisons of different surgical methods for benign prostatic hyperplasia, such as classic transurethral resection of the prostate, enucleation, water vapor ablation, and prostatic artery embolization, etc.) and surgical complications (including major surgical complications, systemic complications, sepsis, sepsis-related encephalopathy, venous thromboembolism and pulmonary embolism, etc.), prognosis and its influencing factors and reproductive effects (including IVF/ICSI treatment), etc.
The research is planned to span 36 months, allowing sufficient time for detailed data analysis, synthesis of findings, and dissemination of results to the scientific community and public.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-the-influencing-factors-of-dementia

Analysis of the influencing factors of dementia

Last updated:
ID:
76636
Start date:
20 October 2021
Project status:
Current
Principal investigator:
Professor Jun Lyu
Lead institution:
Jinan University, China

The occurrence of dementia is slow and insidious, partial dementia patients have a relatively good prognosis if timely detected and early treated, Early diagnosis and prediction of dementia are very important.
Based on the UK population, the aim of our project (estimated duration: 3 years) seeks to investigate the causal relationship between dementia and cardiovascular risk factors, genetic susceptibility, metabolic syndrome and intoxication.
Most of studies focused on the occurrence of dementia induced by risk factors, population researches evaluating how the risk factors affect the development of dementia are limited. This study can further explore the clinical manifestations and influential factors of patients before the onset of dementia, and can provide evidence for the early diagnosis and treatment of dementia in the future.
The potential to shed light on the underlying association between dementia and closely related diseases/risk factors will stress the importance of population-based interventions in the UK and worldwide. Such study will improve the prevention strategies for dementia related diseases and reduce social financial burden on healthcare.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-the-relationship-between-brain-age-and-other-biological-ages-associations-with-health-and-lifestyle-factors

Analysis of the relationship between brain age and other biological ages. Associations with health and lifestyle factors

Last updated:
ID:
916989
Start date:
4 September 2025
Project status:
Current
Principal investigator:
Professor Rodrigo de Luis Garcia
Lead institution:
Universidad de Valladolid, Spain

The aging process does not occur uniformly across biological systems and organs, giving rise to the concept of “biological age” as distinct from chronological age. Among these, “brain age”-a metric derived from neuroimaging data-has emerged as a valuable indicator of brain health and aging. Deviations between brain age and chronological age have been linked to a range of neurological and psychiatric conditions, suggesting that brain age may serve as a useful biomarker for early detection and monitoring of such disorders.

This project aims to advance our understanding of brain age by investigating its relationship with other biological age markers derived from diverse modalities (e.g., ECG, heart MRI, DEXA, full-body MRI). We will explore how discrepancies in brain age correlate with health outcomes, lifestyle choices, metabolic health, cognitive performance, and psychological well-being. In addition, we intend to replicate findings from previous work by our group-conducted in smaller cohorts-regarding the association between brain age and conditions such as migraine, pain-related disorders, and psychotic disorders. A further goal is to explore novel approaches to brain age estimation, including multimodal imaging integration and the evaluation of how image acquisition parameters influence brain age predictions.

Objectives:
* To develop predictive models to calculate brain age and other biological ages obtained from different medical imaging and biomedical signal information.
* To use statistical methods to explore associations between brain age discrepancies and various health conditions, including migraine, pain-related disorders, and psychotic disorders.
* To examine the influence of lifestyle factors, health parameters, and image acquisition parameters on brain age.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-the-relationship-between-lung-function-and-brain-function

Analysis of the relationship between lung function and brain function

Last updated:
ID:
179108
Start date:
17 April 2025
Project status:
Current
Principal investigator:
Rieko Muramatsu
Lead institution:
National Center of Neurology and Psychiatry, Japan

The development of simple and sensitive methods for the assessment of functional brain changes is important for the early detection of brain diseases. However, at present, tough evaluation methods using sophisticated equipment, such as brain imaging, are the mainstay for assessing brain function. It is also difficult to detect slight functional changes in the brain with cognitive function tests. On the other hand, the results of previous basic research have shown that brain function correlates with functional changes in organs other than the brain. Among those peripheral organs, the functional linkage between the lungs and the brain in particular has recently attracted attention. However, it is not known whether changes in brain function can be detected from changes in lung function. This study aims to find a relationship between lung function and brain function from large-scale data. The technology to be developed from the results obtained is expected to detect changes in brain function at an early and acute stage.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-the-uk-biobank-population-prevalence-penetrance-and-effects-of-risk-variants-associated-with-parkinsonism-and-related-movement-disorders

Analysis of the UK Biobank population prevalence, penetrance and effects of risk variants associated with Parkinsonism and related movement disorders

Last updated:
ID:
46450
Start date:
18 June 2019
Project status:
Closed
Principal investigator:
Professor Huw Morris
Lead institution:
University College London, Great Britain

We have identified variants in the genome that increase the risk of neurological illness such as Parkinson’s (LRRK2 G2019S, GBA L444P, and MAPT H1 haplotype). These variants vary in their ability to cause disease; a proportion of variant carriers are unaffected into old age. Our challenge is to understand why some carriers develop disease and others do not, and convert this knowledge into new treatments. Studying these variants in the UK Biobank will allow us to study: 1) their frequency in this population; 2) why some people with these variants get Parkinson’s but some are protected; 3) to comprehensively study the effects of these variants both in terms of disease risk and disease protection. This will enable us to identify how many people might be eligible for a drug study; who is most likely to benefit; if there are any new drug targets and what the potential side effects might be. Our results will enhance our understanding of factors that influence disease risk, and potentially facilitate the development of treatments for illnesses such as Parkinson’s.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-the-uk-biobank-population-prevalence-penetrance-and-effects-of-short-tandem-repeat-expansions

Analysis of the UK Biobank population prevalence, penetrance and effects of short tandem repeat expansions

Last updated:
ID:
351363
Start date:
23 May 2025
Project status:
Current
Principal investigator:
Dr Arianna Tucci
Lead institution:
Queen Mary University of London, Great Britain

Repeat expansion disease are amongst the most common inherited neurological conditions, and collectively affect one in every 3,000 people. These disorders are difficult to assess epidemiologically due to their highly variable clinical presentation, resulting in under-diagnosis.
With the UKBB data, we aim address the difficulty of meaningful REDs frequency estimates and determined the clinical presentation and neuroimaging features of the genetic defect that cause REDs


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-uk-biobank-cohorts-to-accelerate-therapies-to-treat-neurological-disease

Analysis of UK Biobank Cohorts to Accelerate Therapies to Treat Neurological Disease

Last updated:
ID:
93490
Start date:
15 November 2022
Project status:
Current
Principal investigator:
Stefan McDonough
Lead institution:
Atalanta Therapeutics, Inc., United States of America

Ribonucleic acids (RNA) mediate the action of chromosomes in every cell in the body, and the RNA from a specific gene is key in turning that gene’s instructions into actual function. A number of diseases are caused by abnormalities in genes that are turned into abnormal RNA. For example, Huntington’s Disease is caused by lengthening of a specific gene on chromosome 4, and this abnormality is reflected in the corresponding RNA. If one could degrade this pathological RNA throughout the brain, one might arrest Huntington’s Disease. Alternatively, in some neurological diseases there may be health benefit to reducing the expression of normal genes that may be harmful, analogous to genes responsible for high levels of low-density lipoproteins (LDL) that are linked to risk of cardiac disease. The body has a natural physiological process for regulating RNA levels, called RNA interference (RNAi), discovery of which was awarded the Nobel prize in 2006. Artificial RNAi, then, offers the opportunity for specific regulation of individual genes to attack a number of diseases.

Atalanta Therapeutics is a biotechnology company based in Boston, Massachusetts, USA, dedicated to developing therapeutics for neurological and neurodegenerative disease with short interfering RNA (siRNA) technology. Our version of siRNA was engineered to have a selective, durable, and well-tolerated action throughout the brain and central nervous system. Many rare genetic diseases of the nervous system (like Huntington’s) have no effective treatments, and likewise common diseases including Alzheimer’s Disease are in need of truly transformative treatments that attack the root cause of disease. Accordingly, Atalanta’s siRNA technology could be applied to a number of diseases, if key unknowns can be addressed.

The data and infrastructure of the UK BioBank will provide biological understanding of human disease that is key to making actual therapeutics and bringing therapeutics to patients. The UK Biobank will help us select the safest targets for our siRNA, and also help discover any easily measurable biomarkers associated with action of our siRNA that can be used to test the dose at which a potential therapeutic works. The health of persons carrying natural genetic variants in the target of interest will be analyzed, to better understand potential patient populations and to give some measure of the overall prevalence of a specific disease. Results will be made public per UK Biobank policies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-uk-biobank-neuroimaging-data-for-extracting-imaging-based-biomarkers-for-vascular-cognitive-impairment

Analysis of UK Biobank neuroimaging data for extracting imaging-based biomarkers for vascular cognitive impairment

Last updated:
ID:
437499
Start date:
24 March 2025
Project status:
Current
Principal investigator:
Ms Vaanathi Sundaresan
Lead institution:
Indian Institute of Science, India

The project aims are part of DBT-Wellcome Trust India Alliance Early Career Fellowship awarded to the PI.

Research Question:
Can neuroimaging biomarkers for vascular cognitive impairment (VCI) be extracted, and can specific anatomic locations in the brain be identified where the likelihood of these biomarkers increases with various clinical/demographic risk factors (e.g., high blood pressure) in an aging population, thereby validating their utility in the differential diagnosis of VCI?

Project Aims:
This project aims to develop automated, deep learning-based, open-source methods to extract maximal information from neuroimaging data (e.g., lesion count/volume) to build decision-support tools that could assist in the differential diagnosis of VCI.

Proposed methodology:
1. Extraction of Clinically Useful Neuroimaging Biomarkers and IDPs for VCI:
– Utilize semi-supervised learning methods with limited manual annotations.
– Develop privacy-protection techniques (e.g. federated learning) for multi-centre adaptation in real-time.
2. Determining the Clinical Impact of the Spatial Distribution of Biomarkers:
– Study the relationship between the spatial distribution of biomarkers and clinical factors.
– Identify regions with a high probability of biomarker co-occurrence.
– Correlate VCI IDPs with various clinical and demographic factors.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-variants-implicated-in-onset-and-female-bias-in-presentation-of-multiple-sclerosis-to-understand-their-prevalence-and-epistatic-effect-on-multiple-sclerosis-in-the-general-population

Analysis of variants implicated in onset and female-bias in presentation of multiple sclerosis to understand their prevalence and epistatic effect on multiple sclerosis in the general population

Last updated:
ID:
69385
Start date:
26 February 2021
Project status:
Current
Principal investigator:
Dr Kirill Borziak
Lead institution:
Icahn School of Medicine at Mount Sinai, United States of America

The aims of this project is to further characterize alleles that pose a genetic risk for developing multiple sclerosis. A recent family based study has identified a set of alleles that pose a hereditary risk for developing MS. We aim to analyze the prevalence of these alleles in the general population using the UK Biobank dataset to gain better understanding of the importance of these alleles in the onset of MS for unrelated individuals. MS is also aver twice as common in women as it is in men. However, to date very few X-linked alleles have been implicated in the onset of MS and they do not completely explain the increased prevalence of MS in women. This suggests that there are potential other alleles present on the X chromosome which Additionally, we aim to study the interaction between alleles to see using multiple alleles will improve the predictive power of identifying individuals susceptible to MS onset. Previous large scale studies have focused on identifying individual predictive alleles, however, given the variability in the genetic causes of MS susceptibility, we believe a more holistic approach is warranted in identifying the interactions between these alleles.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analysis-of-variants-with-low-minor-allele-frequency-in-the-ukbiobank-cohort

Analysis of variants with low minor allele frequency in the UKBiobank cohort

Last updated:
ID:
55681
Start date:
3 February 2020
Project status:
Closed
Principal investigator:
Dr Jeanette Pruzan Schmidt
Lead institution:
Thermo Fisher Scientific, United States of America

The proposed research aims to validate new genotype calling algorithms for rare variants in array based genotyping data and in particular in the UK Biobank cohort. The results of using the improved algorithms for these variants will be compared to variant information from exon sequencing data available for the UK Biobank cohort. The results will be analyzed by computing both sensitivity as well as positive predictive value of the new algorithms.

Rare variants have been associated with risk factors for many diseases and are therefore of great interest. Several studies have for example examined rare variants in UK Biobank to help explain a person’s risk to develop COPD.

We have characterized the behavior of new genotyping algorithms and have discovered several metrics that appear to significantly improve the genotyping calls of such variants. To date we have applied these new algorithms and metrics to relatively small datasets with known genotypes, such as several hundred HapMap samples. The UK Biobank dataset will allow for the characterization and validation of the algorithms on a large and important dataset.

The results of our research will provide insight on the accuracy of genotype calls using an array platform. Arrays have been shown to provide highly accurate results on variants that are relatively common, (defined as greater than 1% or perhaps 0.1% of the population). The ability to assess the reliability of rarer variants will allow researchers to use array platforms and resulting data for additional applications, such as identifying risk factors for disease.

We expect this project to take less than a year to complete.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analyze-genetic-variants-to-predict-drug-response-and-tailor-medical-treatment-to-an-individuals-genetic-profile-and-identify-frequencies-of-pharmacogenomic-alleles-across-biogeographic-groups

Analyze genetic variants to predict drug response and tailor medical treatment to an individual’s genetic profile, and, identify frequencies of pharmacogenomic alleles across biogeographic groups.

Last updated:
ID:
703474
Start date:
6 June 2025
Project status:
Current
Principal investigator:
Professor Teri E. Klein
Lead institution:
Stanford University, United States of America

Pharmacogenomics (PGx) is an integral part of precision medicine and contributes to the maximization of drug efficacy and reduction of adverse drug event risk. Accurate information on PGx allele frequencies improves the implementation of PGx. We plan to use the resources available with our tool PharmCAT (Pharmacogenomics Clinical Annotation Tool) to extract variants specified in guidelines from a genetic data set derived from sequencing or genotyping technologies, infer the haplotypes and diplotypes, and generate a report containing genotype/diplotype-based annotations and guideline recommendations. The results of these analyses feed directly in our other efforts (PharmGKB: Pharmacogenomics Knowledgebase and CPIC: Clinical Pharmacogenetics Implementation Consortium) which aggregate, integrate and disseminate PGx knowledge and develop and dissemination clinical dosing guidelines which are used world-wide. The ability to identify rare variants and link them to our understanding of the relationship between genotype, environmental factors, and drug response is critical. Additionally, what we currently believe to rare variation, may in fact, not be rare across specific populations. Biobanks with phenotype and drug information are critical to understanding the frequencies of these variants across differing biogeographical groups. Our work will lead to improved understanding of the contributions of drugs to disease progression and will also generate hypotheses about effective treatments.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analyzing-circadian-rhythms-using-multimodal-data-exploring-associations-with-multiple-diseases-and-health

Analyzing Circadian Rhythms Using Multimodal Data: Exploring Associations with Multiple Diseases and Health

Last updated:
ID:
791258
Start date:
9 June 2025
Project status:
Current
Principal investigator:
Professor Rongqian Wu
Lead institution:
The First Affiliated Hospital of Xi'an Jiaotong University, China

Circadian rhythms, governed by core clock genes (e.g., CLOCK-BMAL1) and synchronized by light-dark cycles, regulate systemic physiology, including metabolism, immune response, and cellular repair. Disruptions in these 24-hour cycles are implicated in diverse diseases-including diabetes, cancer, neurodegenerative disorders, and cardiovascular events-with time-dependent “chronorisk windows” driving adverse outcomes. This study aims to systematically investigate population-level variations in circadian rhythms and their associations with diverse disease risks and health outcomes. Leveraging an interdisciplinary approach, we will develop deep learning frameworks to integrate multimodal data sources-including electronic health records (EHRs), environmental exposures, wearable device metrics, and medical imaging-to precisely characterize individual circadian patterns and predict associated health risks.
This research holds transformative implications for understanding circadian mechanisms and their clinical applications. Through multimodal data integration, healthcare providers and individuals may achieve early detection of circadian disruption patterns, enabling precision health strategies that could substantially reduce disease incidence and mortality through timely interventions. By advancing our understanding of the mechanistic links between circadian rhythms and diverse pathologies, this work aims to: (1) elucidate fundamental circadian regulatory principles governing systemic physiology; (2) establish validated circadian biomarkers for disease susceptibility across populations; and (3) inform next-generation predictive models and chronotherapeutic protocols. These insights will catalyze the development of AI-driven tools for personalized circadian monitoring and evidence-based guidelines for optimizing lifestyle/therapeutic timing, ultimately bridging circadian biology to actionable preventive medicine.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analyzing-common-cancers-and-immune-diseases-using-multi-trait-polygenic-risk-score-subtyping-and-imputed-expression-dimensions-for-the-discovery-of-novel-etiological-factors

Analyzing common cancers and immune diseases using multi-trait polygenic risk score subtyping and imputed expression dimensions for the discovery of novel etiological factors.

Last updated:
ID:
62404
Start date:
18 November 2020
Project status:
Current
Principal investigator:
Professor Nicola J Camp
Lead institution:
University of Utah, United States of America

Cancer is an umbrella term. While all cancers are characterized by an excessive proliferation of cells, cancers are usually considered by body site. However, while each cancer-type has similarities, even within each type there are many differences. These differences, or heterogeneities, are due to the many different causes that exist within and across cancer/s. The presence of multiple causes limits our power to detect these causes.
If we could divide patients with a certain cancer-type into subgroups that reflect different underlying etiologies we would increase our understanding of cancer development and potential treatment. Also, if we could add together subgroups of different cancer-types that have shared causes, we would increase power for discovery. These appear somewhat paradoxical, but there is evidence for both. Evidence for subgroups within cancer-types includes observations of differing tumor markers, survival, and responses to therapies. Evidence for shared causes across cancer-types comes from observations of multiple cancers in families and some cancers responding to similar drugs. Immune response has also been offered as a major player in cancer risk. We will define natural clusters based only on genetics across all people in the UKBiobank.
Once these genetic clusters are defined, we will characterize them by the cancers and immune diseases they contain, as well as other health-related, lifestyle, and environmental factors. We have also developed a way to predict the expression of genes for a person based on their genetics. We will use this method to predict gene expression for all people in the UKBiobank, and compare these predicted measures across our genetic clusters. Last, we will look for additional inherited genetic variants that are different across clusters. Through an improved understanding of these clusters, we hope to increase knowledge about cancer-type subgroups (which are split across clusters) and which different subgroups of cancers were clustered together (shared risk) as well as some of the biological relevance of these using gene expression predictions and novel genetic risk factors.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analyzing-disorders-that-cause-optic-nerve-head-swelling-and-vision-loss

Analyzing Disorders That Cause Optic Nerve Head Swelling and Vision Loss

Last updated:
ID:
93071
Start date:
27 September 2023
Project status:
Current
Principal investigator:
Professor Mark J Kupersmith
Lead institution:
Icahn School of Medicine at Mount Sinai, United States of America

Neurologic and systemic diseases contribute to or cause swelling of the optic nerve head and vision loss. These disorders include optic neuritis related to multiple sclerosis, papilledema due to idiopathic intracranial hypertension (IIH) and cardiovascular risk factor associated non-arteritic anterior ischemic optic neuropathy (NAION). Visual field testing, optical coherence tomography (OCT) and fundus photos are used to diagnose, monitor, and measure the outcome and effects of therapies. Each disorder appears to have distinct features of ONH swelling, injury, vision loss, and recovery. We are developing artificial intelligence (AI) methods to uncover these features. We hypothesis that AI will identify specific photographic or OCT or both features that will be biomarkers that change over time due to reflect the natural history, response to therapy, and/or predict swelling or visual outcome. We suggest that these ONH disorders will be distinguishable based on these features and improve diagnostic capabilities.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analyzing-genotype-phenotype-associations-with-fairness-and-privacy

Analyzing genotype-phenotype associations with fairness and privacy

Last updated:
ID:
93997
Start date:
28 March 2023
Project status:
Closed
Principal investigator:
Professor Seth Neel
Lead institution:
Harvard University, United States of America

The goal of this project is to conduct a comprehensive study of fairness and privacy of sharing summary statistics about genomic datasets, in light of significant advances in the field of algorithmic fairness and data privacy over the last 10 years. It is our hope that new tools can be applied within the genomic context to unlock high-fidelity sharing of the results of genomic analyses that can improve health outcomes not just for a specific population, but broadly for diverse groups of individuals of different ancestry and sociodemographic backgrounds. This has the potential to unlock greater prediction accuracy for clinical insights, collaboration, data access, and statistical power, across all studies using genomic data — both safeguarding data sharing in a more rigorous way and unlocking future discoveries.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analyzing-mendelian-disease-related-phenotypic-variability-using-population-scale-data

Analyzing Mendelian Disease-related Phenotypic Variability using Population-scale Data

Last updated:
ID:
99922
Start date:
17 April 2023
Project status:
Current
Principal investigator:
Dr David Randall Blair
Lead institution:
University of California, San Francisco, United States of America

DNA is like an instruction booklet for our bodies. It contains sentences, words, and single letters. This booklet is passed down from parents to children, and it shapes how our bodies grow and develop. Each booklet contains slightly different letters. Most of these genetic differences do not impact our health, but some cause serious problems. In fact, scientific studies have shown that nearly 1% of all people suffer from a disease that’s caused by only one or two DNA changes. These are often called Mendelian diseases. Ideally, we would test everyone for the DNA changes that cause Mendelian diseases right after they are born. This way, we would immediately know who is at risk for these disorders. Unfortunately, this is more difficult than it sounds. Some people with DNA changes linked to Mendelian diseases never develop symptoms, and if they do, the symptoms are mild and don’t start until adulthood. If we simply tested everyone for all possible Mendelian disease DNA changes at birth, then we would find many people with suspicious findings that would never develop severe symptoms. This would lead to lots of unnecessary testing, treatments, and stress for these patients and their families. As a result, testing everyone for the DNA changes that can cause Mendelian diseases is impractical.

In this project, we plan to study the DNA changes that cause Mendelian diseases in greater detail using the UK Biobank. Specifically, our goal is to determine why some people with these changes develop severe symptoms while others seem relatively unaffected. To accomplish this goal, we will first measure the severity of different Mendelian disease symptoms using all the subjects in the UK Biobank. After combining this information with genetic data, we hope to find both severely and mildly affected subjects that carry DNA changes associated with these diseases. By comparing these two groups of people, we plan to identify different factors that predict whether a patient will experience mild or severe symptoms. Examples of such factors include other DNA changes, medications, and environmental exposures like smoking. If successful, our project will provide new information about why people with the same or similar DNA changes develop different symptoms. This will help us provide more personalized healthcare to patients impacted by these disorders. In addition, this information should help us perform DNA testing for Mendelian diseases in more patients, particularly those who have yet to develop symptoms.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analyzing-minor-allele-homozygosity-patterns-and-correlations

Analyzing Minor Allele Homozygosity: Patterns and Correlations

Last updated:
ID:
457958
Start date:
6 May 2025
Project status:
Current
Principal investigator:
Mr Yitzhak Pilpel
Lead institution:
Weizmann Institute of Science, Israel

We aim to analyze minor allele homozygosity across different populations using the UK Biobank dataset. Our research seeks to answer several key questions:
1. How do homozygosity levels vary across populations?
2. Do certain minor alleles become more frequent, and do these changes correlate with environmental factors?


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analyzing-mitochondrial-and-nuclear-genome-variation-to-improve-variant-interpretation

Analyzing mitochondrial and nuclear genome variation to improve variant interpretation

Last updated:
ID:
977700
Start date:
30 October 2025
Project status:
Current
Principal investigator:
Dr Nicole Lake
Lead institution:
Yale University, United States of America

This project will investigate how genetic variation in the human genome, with a focus on the mitochondrial genome, contributes to health and disease. We aim to characterize patterns of variation in mitochondrial DNA (mtDNA), quantify constraint across the mitochondrial genome, and use these and related models to interpret the functional and clinical relevance of observed variants. We will also explore how mitochondrial variants interact with variation in the nuclear genome to shape molecular and physiological traits.
Key research questions include: (i) which regions of the mitochondrial genome are most intolerant to variation, and how does this relate to known or predicted variant effects; (ii) how can measures of evolutionary constraint and variant deleteriousness be integrated to improve interpretation of mitochondrial variants; (iii) how combinations of mitochondrial and nuclear variants contribute to differences in phenotypes and disease risk.
Our objectives are to: (1) Develop next generation models of mitochondrial constraint using population-scale variation data; (2) Analyze patterns of rare and common variation to identify potentially deleterious variants; (3) Assess how variation in mtDNA and nuclear genes influence phenotypes and disease outcomes; (4) Generate broadly applicable variant interpretation resources for research and clinical use.
The scientific rationale is that mtDNA remains underutilized in large-scale human genetics, and integrating statistical models, genome-wide data, and phenotype information will advance our understanding of variant effect and disease mechanisms.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analyzing-the-genetic-association-between-depressive-disorders-and-metabolic-conditions-considering-environmental-risk-factors

Analyzing the genetic association between depressive disorders and metabolic conditions considering environmental risk factors

Last updated:
ID:
61700
Start date:
29 September 2020
Project status:
Closed
Principal investigator:
Professor Hans Grabe
Lead institution:
University Medicine Greifswald, Germany

Studies have shown that persons with metabolic alterations are more likely to develop depressive symptoms or even a depressive disorder. At the same time the risk of developing metabolic disorders is higher in individuals with depressive symptoms. Both conditions have increasing prevalence worldwide. However, the reasons for these frequent co-occurrences are not yet fully understood. The conditions may be direct consequences of each other or impacted by common genetic or environmental risk factors such as trauma, education, social status or behavioral traits. By combining genetic and environmental/lifestyle data in our analyses we aim to get a better medical understanding of the factors underlying this comorbidity. Results of our project will have the potential to improve early recognition of individuals who are at risk for developing depressive disorders or metabolic diseases. Moreover, the findings might help developing improved preventative and treatment approaches for people with both conditions. Furthermore, a better understanding of depressive disorders may reduce stigma toward depression in the general population and increase help seeking. Our project will be conducted over an extended period of approximately 3 years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analyzing-the-relationship-between-physical-activity-and-cognitive-brain-health-after-cancer

Analyzing the Relationship between Physical Activity and Cognitive & Brain Health after Cancer

Last updated:
ID:
71363
Start date:
3 November 2021
Project status:
Current
Principal investigator:
Dr Elizabeth Ann Salerno
Lead institution:
Washington University in St. Louis, United States of America

Deaths from cancer have declined significantly over the past several decades. However, a diagnosis of cancer and its treatment are not without their consequences. Cancer survivors often struggle with a host of physical and mental concerns after treatment has ended, one of which is cognitive impairment, or “chemo brain.” Chemo brain has been shown to have significantly negative economic, social, and functional implications as survivors as it can prevent survivors’ from readily returning to their normal daily activities. It is crucial that we identify strategies for treating chemo brain, thereby improving cancer survivors’ quality of life.

Physical activity is a lifestyle behavior that has been consistently associated with improved health in cancer survivors. Preliminary research has suggested that it might help chemo brain, but this work has been conducted in small samples of homogeneous survivors and is not representative of the entire population of cancer survivors. We know little about the relationship between physical activity and the brain in young vs. old, white vs. black vs. latinx, or rural vs. urban survivors. These factors have been important in other areas of cancer survivorship and may represent key targets for future interventions. Understanding these relationships is critical if we hope to design physical activity programs for specific groups of survivors.

Our overarching research question is to better understand the relationships between physical activity, sedentary behavior, and cognitive and brain health during long-term cancer survivorship and how this association may vary across diverse groups of cancer survivors. Accordingly, our aims are as follows:
1. Quantify the association between physical activity and cognitive and brain health in individuals previously diagnosed with cancer. We hypothesize that survivors who are more active (e.g., more moderate-to-vigorous, light, and total physical activity) will demonstrate preserved brain structure (e.g., greater hippocampal volume and white matter integrity), function (e.g., resting state functional connectivity in several networks), and cognitive functioning (e.g., better executive functioning, faster processing speed).
2. Quantify the relationship between sedentary behavior and cognitive and brain health in those with a history of cancer. We hypothesize that more sedentary survivors will demonstrate more deteriorated brain structure and function and poorer cognitive functioning.
3. Explore these associations within different strata of cancer survivors (e.g., age, race, environment). We hypothesize that older survivors, survivors of color (i.e., Black), rural-dwelling survivors, and those who received chemotherapy will demonstrate more deteriorated brain structure and function and poorer cognitive functioning.
Estimated duration: 24 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/analyzing-unmappable-camouflaged-genome-regions-in-uk-biobank-exome-sequencing-data-using-the-lpa-gene-as-a-model

Analyzing unmappable (“camouflaged”) genome regions in UK Biobank exome sequencing data using the LPA gene as a model.

Last updated:
ID:
62905
Start date:
27 July 2020
Project status:
Current
Principal investigator:
Professor Sebastian Schoenherr
Lead institution:
Medical University of Innsbruck, Austria

The human genome includes still thousands of regions that cannot be properly analysed (called camouflaged or dark regions). The affected genes belong to pathways important to human health, development and reproduction and represent nearly one third of all human protein-coding genes.
In this project, we will analyze in depth such a region, which has also a very large impact on human health, the LPA gene. This gene regulates the Lipoprotein(a) [Lp(a)] concentrations, which are among the strongest genetic risk factors for cardiovascular diseases, myocardial infarction and aortic valve calcification. Moreover, they have been implicated as a risk factor for type 2 diabetes. Lp(a) levels are regulated nearly exclusively genetically by genetic variation in the LPA gene but most of the gene is located in a hitherto not analyzable region named the “KIV-2 repeat”. Therefore despite its tremendous importance of LPA for cardiovascular disease and human health little is known about how genetic variation in LPA regulates Lp(a) concentrations in detail. Indeed, about one fifth of the general population (>100 million people in Europe) presents genetically increased Lp(a) levels and thus a better understanding of the mechanisms that regulate Lp(a) can provide a tremendous impact on public health.
In this project we will apply to this gene new bioinformatic and statistical approaches that we have developed for previous smaller studies. We aim at evaluating our approaches to (1) search for novel variants in the LPA gene that influence Lp(a) concentrations, cardiovascular risk and human health, (2) expand our approach also to other clinically camouflaged protein-coding regions and (3) finally provide the scientific community with a computational pipeline to efficiently analyse such complex regions in very large studies like UK Biobank. We expect a time frame of 3 years to provide results to the community.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/ancestral-adaptation-nutrient-intake-and-metabolic-disease-physiological-modeling-and-econometric-testing

Ancestral Adaptation, Nutrient Intake, and Metabolic Disease: Physiological Modeling and Econometric Testing

Last updated:
ID:
983973
Start date:
29 October 2025
Project status:
Current
Principal investigator:
Professor Brandon Ogbunugafor
Lead institution:
Yale University, United States of America

This project investigates the hypothesis that nutrient intake contributes to metabolic disease (hypertension and diabetes) but only when it exceeds person-specific thresholds, shaped by ancestral adaptation to resource scarcity.
Hypertension: We hypothesize that salt intake raises arterial pressure and contributes to cardiovascular disease (CVD), but only beyond an individual threshold. This threshold-and an associated arterial pressure set point-is traced back to ancestral sodium availability.
Objectives:

(i) Develop a physiological model incorporating the threshold mechanism.
(ii) Examine the model’s implications for the arterial pressure-sodium intake and CVD-sodium intake associations.
(iii) Validate the mechanism by testing whether CVD increases with sodium intake, but only in salt-sensitive individuals.
Diabetes: We hypothesize that caloric intake raises BMI and contributes to diabetes, but only beyond a person-specific threshold that is determined by ancestral caloric availability.
Objectives:
(i) Build a physiological model of diabetes that incorporates this threshold.
(ii) Test the model’s implications for BMI-caloric intake and diabetes-caloric intake associations.
(iii) Validate the mechanism by checking whether diabetes risk increases with caloric intake, but only for individuals who have crossed their threshold.

Scientific Rationale:
Conflicting findings on the link between sodium intake, arterial pressure, and CVD are explained by heterogeneity in sodium sensitivity in our model, which arises due to underlying person-specific set points. This implies nonlinear (discontinuous) sodium intake-arterial pressure-CVD associations that we will test. Variability in the BMI-diabetes association across individuals and ethnic groups is explained in our model by heterogeneity in BMI set points, rather than by other risk factors. This implies nonlinear (discontinuous) caloric intake-BMI-diabetes associations that we will test.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/angioedema-investigation-of-genetic-and-environmental-risk-factors

Angioedema: Investigation of genetic and environmental risk factors

Last updated:
ID:
60928
Start date:
10 July 2020
Project status:
Closed
Principal investigator:
Professor Markus Nöthen
Lead institution:
University of Bonn, Germany

The aim of the present research project is to investigate the molecular basis of bradykinin-mediated tissue swellings (angioedema) with a focus on angiotensin converting enzyme inhibitor (ACEi)/ angiotensin receptor blocker (ARB) induced angioedema and hereditary angioedema (HAE). Angioedema can become life-threatening events if, e.g., the tongue, larynx or throat are affected. They can appear as a known side effect of ACEi and ARBs. Based on the results of previous research it is assumed that susceptibility to ACEi/ARB-induced angioedema is dependent on a genetic predisposition as well as personal and environmental risk factors. However, the exact biological mechanisms contributing to these angioedema are still largely unknown. Another type of bradykinin-mediated angioedema is HAE which is a rare genetic disease associated with recurrent episodes of tissue swellings. To date, genetic mutations have been identified in four different genes being causal for around 75% of HAE cases. However, in the remaining proportion of patients, the underlying genetic cause is still unknown, which often makes diagnosis and individual therapy difficult or delayed. In addition, not all patients with pathogenic HAE mutations show clinical symptoms. To improve prevention, diagnosis and patient outcome in ACEi/ARB-induced angioedema and HAE, we will use the UK Biobank data and perform state-of-the-art molecular genetic analyses (including genome-wide association and exome sequencing analyses) to (i) identify genetic and non-genetic risk factors for ACEi/ARB-induced angioedema, (ii) assess the frequencies of known monogenic HAE forms within the general population, and (iii) identify potential new candidate genes for HAE. Our results could lead to a better prediction of the occurrence of such angioedema and enable new preventive, diagnostic and therapeutic approaches.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/antenatal-selection-and-fetal-health

Antenatal selection and fetal health

Last updated:
ID:
176618
Start date:
11 November 2024
Project status:
Current
Principal investigator:
Ms Ramina Sotoudeh
Lead institution:
Yale University, United States of America

A lot of public and scholarly attention has been paid to improving infant health and reducing infant mortality worldwide. However, the role and mechanisms through which prenatal conditions affect infant health are not adequately examined. Scholarly literature has shown that the uterine environment becomes more adverse for fetuses as the mother ages. As our societies are moving towards later ages at birth maternal age, this question has become even more pressing. Although our question is concerned with prenatal conditions, we use a population who survived to adulthood, in order to answer it. We will look at the genetics of birth order and the differences in genotypes across siblings (who were exposed to different prenatal exposures) as our methodological strategy. This research will shed light not only on the conditions of infants after birth, but also on which embryos survive to term, and thus the composition of the population in a given birth cohort. These results will have implications for the kinds of interventions, especially targeted towards prenatal conditions of both parents, that would be effective.

The anticipated duration of the project is about 3 years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/anthropometric-characteristics-and-the-risk-of-atrial-fibrillation-and-cardiovascular-diseases-in-different-age-groups

Anthropometric characteristics and the risk of atrial fibrillation and cardiovascular diseases in different age groups

Last updated:
ID:
76593
Start date:
8 February 2022
Project status:
Current
Principal investigator:
Dr Eue-Keun Choi
Lead institution:
Seoul National University Hospital, Korea (South)

It is important to identify factors that have causal relationship with cardiovascular events and can be targeted for treatment. Using large-scaled individual data from the UK Biobank, this study aimed to focus on the data of anthropometric measurements such as body weight, waist circumference, and so on, along with whole-body bio-impedance analysis results indicating the quantitative data of body composition (for example, fat percentage and mass, muscle mass).

The present project will last for a total of 3 years. This study is expected to identify important body measurement and body composition parameters that are important in accurately predicting individual cardiovascular outcomes, with a particular emphasis on atrial fibrillation. We will investigate the relationship between the parameters and the illnesses, which may lead to the identification of specific targets for treatment and prevention of cardiovascular diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/antibiotic-exposure-and-life-history-outcomes

Antibiotic Exposure and Life History Outcomes

Last updated:
ID:
75157
Start date:
24 February 2022
Project status:
Closed
Principal investigator:
Professor Jay Stock
Lead institution:
University of Western Ontario, Canada

This research study examines how changes in human-microbial interactions in childhood can impact growth and health outcomes in later-life. By using antibiotic’s known ability to disrupt microbes’ function, this study uses antibiotics’ disruptive ability to examine the influences microbes can have on growth and disease outcomes.
The aim of this research is to improve understanding of how microbes influence life in short- and long-term contexts in addition to how early-life influences can produce effects later in life. Understanding these interactions is complex but by controlling for factors which have known influences on human microbes (e.g. diet and medications), we can test for trends that indicate microbially-influenced growth and health outcomes. These trends can then be used to examine changes in historical growth and health patterns which currently lack robust explanations. For modern populations, this study provides a detailed long-term examination of childhood antibiotic influence on later-life health outcomes which could inform the broader understanding of the consequences of antibiotic use.
This research will be conducted over the course of twelve months with the bulk of research being published in a Master’s thesis. Additionally, results that warrant further exploration will be published in academia journals and public outreach programs with all information regarding data use and publication being sent to the UK Biobank.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/antibiotic-use-and-dementia-risk-or-cognitive-change

Antibiotic use and dementia risk or cognitive change

Last updated:
ID:
57999
Start date:
30 April 2020
Project status:
Closed
Principal investigator:
Dr Zhou Liu
Lead institution:
Affiliated Hospital of Guangdong Medical University, China

Gut microbiota dysbiosis are likely to be involved in the development of dementia. Antibiotics is one of the main cause of dysbiosis. However, some dementia animal studies show that antibiotic is beneficial. And the associations between antibiotics and cognition decline or dementia remain unclear.

Therefore, our objective is to investigate the associations between antibiotic use and cognition decline or dementia risk in the large, prospective UK Biobank study.

To assess these associations, we will use statistical models which take into account variables that may affect cognition and dementia, such as education, income, hypertension, diabetes, and gene. These findings can provide evidence and further understand the role of antibiotics in dementia, and provide evidence for continued exploration of antibiotic use. Findings from this study may also have major implications for prescribing practices and public health.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/antibodies-to-human-herpesviruses-and-risk-of-incident-cardiovascular-events-and-all-cause-mortality

Antibodies to human herpesviruses and risk of incident cardiovascular events and all-cause mortality

Last updated:
ID:
63816
Start date:
7 August 2020
Project status:
Closed
Principal investigator:
Professor Charlotte Warren-Gash
Lead institution:
London School of Hygiene and Tropical Medicine, Great Britain

Herpesviruses are common: around 95% of UK adults have been infected with varicella zoster virus and around 50-70% have experienced herpes simplex virus type 1 or cytomegalovirus infection. After initial infection, these viruses lie dormant, but they can reactivate when the immune system is under stress. Some studies have linked virus reactivation to other diseases such as stroke and dementia. However, a better understanding of the nature of these relationships is essential to identify who is at risk and during which time periods, to guide preventive efforts.

Research into herpesviruses is hampered by difficulties measuring virus reactivation. While some people experience typical signs and symptoms such as cold sores (due to herpes simplex virus), or shingles (a painful blistering rash caused by varicella zoster virus), these events may not be well recorded in GP or hospital records. Antibodies against the viruses detected in blood samples show a person’s infection status, while the level of antibodies may reflect the degree of virus reactivation.
In this study, we will investigate whether having high antibody levels to three common herpesviruses is associated with the risk of heart attack, stroke and death in the UK Biobank cohort. This will inform future research into interventions such as vaccines to prevent complications of herpesviruses in older age.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/antidepressant-use-and-risk-of-morbidity-and-mortality-a-population-based-cohort-study

Antidepressant use and risk of morbidity and mortality: a population-based cohort study

Last updated:
ID:
46704
Start date:
30 August 2019
Project status:
Closed
Principal investigator:
Dr Narinder Bansal
Lead institution:
University of Bristol, Great Britain

Antidepressant prescribing has risen dramatically over the past decade, mainly because patients are being prescribed these drugs for a longer period. Some patients may end up taking these drugs for longer than they would like because of concerns that the depression may recur if they stop, and because of concerns relating to withdrawal symptoms. Many patients (nearly half according to a Scottish study) are staying on these drugs for more than two years. Given the rise in longer treatment, it is important to find out whether there are any negative health effects associated with taking these drugs so that doctors and patients can discuss these when making prescribing decisions. The public health impact of this work is high given the increasing number of antidepressants prescribed in England (61 million prescriptions in 2015) and the lack of research on long-term safety. Some of the common symptoms reported by patients suggest that these drugs may affect the cardiovascular system, liver and brain. Recently, two large studies, using data gathered from GP practices (covering 11 to 12 million patients) have shown that antidepressants increase the risk of conditions such as epilepsy, fractures and dementia. Many patients who start antidepressants are already at risk of developing these conditions. We want to find out the extent to which taking an antidepressant adds to this risk. To assess the size of this risk more accurately, we need to take into account a person’s risk for this disease or condition before they started taking antidepressants. To do this, we need detailed information on key risk factors for all the diseases and conditions of interest. Unfortunately, previous studies have been unable to take these risk factors into account because this information is not measured and recorded for everyone in primary care. We plan to use data collected by UK Biobank, a large study that recruited just over 500,000 participants from across the UK between 2006 to 2010. This study is an ideal resource to explore the long-term effects of antidepressants because it has extensive information on a range of risk factors for disease and it is also linked to GP and hospital records allowing us to look at the effect of antidepressants on a wide range of conditions and diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/aortic-elongation-and-stiffness-insights-from-biobanks

Aortic elongation and stiffness – Insights from biobanks

Last updated:
ID:
81032
Start date:
9 September 2022
Project status:
Current
Principal investigator:
Professor Julio Alonso Chirinos
Lead institution:
University of Pennsylvania, United States of America

The aorta is the largest artery (pipe) going out of the heart. Young, healthy non-stiff aortas accommodate and “cushion” the intermittent cardiac ejection and transform it into steady flow at the level of the microvasculature (tiny vessels that feed our various organs). As individuals age, stiffening and deformation of the aorta hinders this process. About half of the variability in aortic stiffness is heritable, suggesting important genetic components. We are developing a new method to measure aortic stiffness using echocardiography, a widely available tool. This requires a validated equation to estimate the length of the aorta. Moreover, the aortic length is important in and of itself because the aorta lengthens with age and disease. Aortic lengthening is thus a key parameter of aortic aging, but its determinants are poorly understood. In fact, its genetic determinants have never been investigated. To address these important gaps, we will pursue the following aims:
1) To characterize aortic elongation with age and its genetic determinants, as well as its relationship with cardiovascular risk and blood pressure
2) To develop a widely applicable equation to estimate aortic length, derived from the UK biobank
We expect this project to take 3-4 years to complete.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/aortic-stiffness-and-the-pathogenesis-of-cardiovascular-disease

Aortic stiffness and the pathogenesis of cardiovascular disease

Last updated:
ID:
88279
Start date:
13 May 2022
Project status:
Current
Principal investigator:
Dr Gary Frank Mitchell
Lead institution:
Cardiovascular Engineering, Inc., United States of America

When the heart beats, it pumps blood into the arterial system. The heart is connected directly to the aorta, which is the largest artery in the body. The normal aorta is highly elastic and buffers the pressure swings that accompany each heartbeat. When the aorta stiffens, the pressure swings in the aorta and arteries throughout the body increase and can lead to development of high blood pressure. These abnormal swings in blood pressure during each heartbeat can damage fragile small vessels in high flow organs such as the brain and kidneys, leading to cognitive impairment, dementia and chronic kidney disease. In addition, the excessive pressure swings increase load on the heart and can result in enlargement of the heart and development of heart failure. Our research project will define factors that contribute to stiffening of the aorta and will examine consequences of aortic stiffening on brain, kidney and heart structure and function. Identification of factors that contribute to stiffening of the aorta will offer insight into potential lifestyle modifications or treatments that can prevent or reverse aortic stiffening and limit the premature morbidity and deaths attributable to aortic stiffening and high blood pressure.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/apoe-genotype-as-an-effect-modifier-of-the-impact-of-diet-on-cognitive-health-outcomes-and-all-cause-mortality

APOE genotype as an effect modifier of the impact of diet on cognitive health outcomes and all-cause mortality.

Last updated:
ID:
988999
Start date:
28 September 2025
Project status:
Current
Principal investigator:
Dr Jakob Norgren
Lead institution:
Karolinska Institutet, Sweden

Apolipoprotein E genotype (APOE) is the predominant risk modifier for Alzheimer’s disease. Empirical evidence published by us and others support the hypothesis that APOE modifies the response to dietary factors, based on evolutionary adaptation. These findings warrant replication. Our aim is to estimate the effect of compositional dietary factors on cognitive health outcomes and mortality. Primarily, we will dichotomize APOE 3/4 and 4/4 (APOE34/44) versus non-APOE34/44. We will also examine the six common genotypes separately, to explore the hypothesis that effect modification will follow a gradient: APOE22-23-24-33-34-44.

Primary exposure: Unprocessed meat (by weight, standardized for total energy intake). Secondary exposures: Total meat and ratios between meat types. Other food groups (egg, fish, seafood, tubers, nuts, vegetables, legumes, cereals, dairy etc.); Macronutrient parameters: Carbohydrates/Fat (log-ratio), and protein E(%); Micronutrient parameters: vitamin B12 etc..; Metabolic biomarkers

Future directions of the project may include other health and disease outcomes. The exposures of interest may be extended to medications, particularly lipid lowering drugs, hypertensives, and diabetes drugs. These extensions will typically target the overall research question of APOE as an effect modifier, potentially extending to other genes of interest.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/apoe-lifestyle-interactions-on-age-related-health-outcomes-in-the-uk-biobank-population

APOE-lifestyle interactions on age-related health outcomes in the UK Biobank population

Last updated:
ID:
32292
Start date:
2 July 2018
Project status:
Closed
Principal investigator:
Dr Raymond Noordam
Lead institution:
Leiden University Medical Centre, Netherlands

In genome-wide association studies, the APOE gene was associated with multiple aging-related phenotypes (notably Alzheimers disease, cardiovascular disease and mortality). There is increasing evidence in literature that suggests that lifestyle factors (e.g., food intake, physical activity) interact with the variation in the APOE gene in the development of multiple age-related diseases. These interactions are potential promising strategies to prevent or delay disease onset. However, these studies were generally small in total sample size. Therefore, we aim to investigate APOE-lifestyle interactions in relation to age-related diseases in the UK Biobank. The UK Biobank aims to improve prevention, treatment and diagnosis of several life-threating illnesses throughout society. The proposed research will add to a better understanding of the interaction of variation in the APOE gene with lifestyle factors on multiple age-related diseases and mortality. This research aims to add evidence whether there is any need to specifically target APOE risk allele carriers for first-line preventive strategies. This research project may contribute to decrease and delay the development of disease in APOE-risk carriers, in order to improve quality of life and to decrease disease burden and mortality. In the present study, we aim to assess whether the association between APOE genetic variants and age-related diseases (cognition, depression, diabetes and cardiovascular disease) and mortality is modified by different lifestyle factors (notably objectively collected physical activity, nutrition, sleep, alcohol intake and smoking). For the present proposal we will use all participants of the UK Biobank with genotyped data (~500,000 participants maximum; ~100,000 participants with objectively collected physical activity data; ~211.000 participants with diet by 24-hour recall data).


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/apogee-polygenic-architecture-of-adult-morbid-obesity-in-european-populations

APOGEE – Polygenic Architecture of Adult Morbid Obesity in European Populations

Last updated:
ID:
135387
Start date:
30 April 2024
Project status:
Current
Principal investigator:
Dr Sébastien Hergalant
Lead institution:
INSERM, France

In constant rise in developing and developed countries, obesity, defined as an abnormal or excessive accumulation of fat that presents a risk to health, represents a worldwide and public health concern. Obesity is a multifactorial disease resulting from the interaction of genetic and environmental factors. To date, only 7 polymorphisms have been associated with the risk of morbid obesity in populations of adult European ancestry. Body mass index (BMI) is the parameter most frequently used to diagnose obesity in the population.
This study aims to explore the genome of > 400,000 adults of normal weight and > 70,000 adults with morbid obesity (BMI ! 40 kg/m2) by meta-analysing multiple large cohorts of European ethnicity, to discover new genetic variants predisposing to obesity, to explain BMI variations in the different obese categories, and apply these results to the general population. For up to 3 years, we will highlight the polygenic architecture of morbid obesity, perform genome-wide association studies (GWAS) meta-analyses, score regression analyses and post-GWAS augmentations with state of-the-art bioinformatics approaches to decipher biological functions and pathways linked to morbid obesity.
Current obesity treatment programs however have largely failed to halt the obesity epidemic, despite huge financial investments that can represent up to 10% of national health expenses. Exploring the causes and consequences of obesity might improve prediction, prevention and care in the long run. This project holds the potential for discovering new genes predisposing to obesity, decipher the complex gene network underlying its physiopathology, understand the molecular links to its comorbidities (cardiometabolic diseases, cancers, type 2 diabetes), and identify novel molecular targets for innovative medications.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/apol1-risk-variants-and-the-risk-of-chronic-kidney-disease-in-a-multi-ethnic-population

APOL1 risk variants and the risk of chronic kidney disease in a multi-ethnic population

Last updated:
ID:
914676
Start date:
4 September 2025
Project status:
Current
Principal investigator:
Ms Emma van Schijndel
Lead institution:
Amsterdam UMC Research BV, Netherlands

The apolipoprotein L1 gene (APOL1) contains two well-known variants, G1 and G2, which are linked to an increased risk of kidney disease. These risk variants are almost exclusively found in individuals of Sub Saharan West African descent. Initially, it was believed that a person needed to carry two risk variants along with a ‘second-hit’ to be at increased risk of kidney disease. However, recent studies from Ghana and Nigeria suggest that the risk may follow an additive pattern. This was shortly thereafter supported by a study from the United States of America involving individuals with HIV.
This study aims to validate the evidence we found for an additive risk model in the multi-ethnic HELIUS cohort from Amsterdam. We will analyse the association between APOL1 risk variants and various measures of kidney function, adjusting for traditional risk factors for kidney disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/appetite-and-its-role-as-a-causal-determinant-of-binge-eating-and-related-mental-health-disorders

Appetite and its role as a causal determinant of binge eating and related mental health disorders.

Last updated:
ID:
826297
Start date:
7 October 2025
Project status:
Current
Principal investigator:
Dr Ilaria Costantini
Lead institution:
University College London, Great Britain

Binge eating disorder (BED) is the most common eating disorder and is associated with high mortality and morbidity. Despite its prevalence and severity, BED is under-researched, under-recognised, and under-treated. Dysregulated levels of appetite-regulating hormones (e.g., ghrelin, GHRL; glucagon-like peptide-1, GLP1) are putative risk factors for binge eating, a core feature of BED. These hormones may act via biological mechanisms – influencing brain regions involved in homeostatic regulation, reward processing, and inhibitory control – or environmental pathways – such as weight stigma linked to increased weight gain, and consequent cycles of restriction and binge eating. Drugs targeting these hormones, such as GLP1 receptor agonists (GLP1RAs, e.g., semaglutide), could hold potential as adjunct treatments for binge eating.
This project aims to understand the genetic and environmental causal determinants of binge eating, with a particular focus on appetite and its biological and environmental downstream mechanisms. This study aims to obtain novel mechanistic insights into binge eating risk, identify susceptible individuals, and establish preventative and therapeutic targets through analysis of risk factor associations using novel causal inference methodology. Causal inference methods such as PRS analysis, Mendelian randomization (MR) analyses will be used alongside traditional epidemiological methods such as moderation and mediation analyses. MR and PRS analyses will consider any potential risk factor, including (but not limited to) anthropometric (e.g., adiposity), social (e.g., weight-stigma), and molecular markers (e.g., metabolites, proteins). For example, we will use GRS for each drug target to assess whether the genetic relationship of appetite-regulating hormones with binge eating varies according to an individual’s genetic liability to higher weight.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/applicability-of-the-reduce-it-trial-to-uk-biobank

Applicability of the REDUCE-IT trial to UK Biobank

Last updated:
ID:
734824
Start date:
21 October 2025
Project status:
Current
Principal investigator:
Ms Marlo Verket
Lead institution:
Uniklinik RWTH Aachen, Germany

The REDUCE-IT (Reduction of Cardiovascular Events with Icosapent Ethyl-Intervention Trial) demonstrated a significant reduction in atherosclerotic cardiovascular disease (ASCVD) risk using a treatment strategy of high-dose omega-3 icosapent ethyl compared to placebo in statin-treated patients with elevated triglycerides (150 mg/dL or more) and well-controlled low-density lipoprotein cholesterol (LDL-C). There was a 25% cardiovascular (CV) risk reduction (composite of cardiovascular death, nonfatal myocardial infarction, nonfatal stroke, coronary revascularization, or unstable angina) in those who have received the high-dose omega-3 icosapent ethyl in comparison to those received placebo. One limitation of randomized trials is that their populations may not accurately reflect those treated in everyday clinical practice.
Therefore, we would like to use UK Biobank to select populations that included ASCVD patients who are on statin therapy. Then, we will split the groups up to patients who reflects the REDUCE-IT inclusion and exclusion criteria with or without diabetes and those who do not reflect the trial. Additionally, we will split the groups based on triglycerides into three groups – normal, elevated and very elevated. With these groups, we will evaluate the characteristics, management and cardiovascular outcomes, such as MACE, death, hospitalisation due to heart failure in the UK individiuals. We will calculate the event rate and odds ratio, using SPSS. Then, we will compare this to results of REDUCE-IT to identify the applicability or differences in results from a clincial trial like REDUCE-IT and observational data, UK Biobank. This will hopefully identify the unmet medical need to find other therapeutic strategies to help reduce the residual risk of cardiovascular outcomes in patients with high triglycerides.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/application-of-a-deep-learning-system-for-detecting-prediabetes-based-on-eye-image-and-life-style

Application of a deep learning system for detecting prediabetes based on eye image and life style

Last updated:
ID:
49490
Start date:
28 February 2020
Project status:
Closed
Principal investigator:
Dr Wei Wang
Lead institution:
Sun Yat-Sen University, China

Diabetes mellitus (DM) is one of the most graving public health challenges worldwide that brings enormous harm and burden to patients and societies. Due to its complicated diagnostic methods and limited screening manners, the detection rate of patients with early diabetes mellitus is very low worldwide. As the deep learning algorithms that feature using image recognition to extract characteristics mature, early artificial intelligence(AI)-based diabetes mellitus screening model has been initially established. However, external clinical validation in population of different ethnicities is still lacking. In this study, we hope to use the color fundus images and lifestyle-related variables (such as smoking, drinking, and exercising) in the database to validate the established AI-based model. The results will prove the validity of the model, which will help improve not only the screening detection rate of early diabetes mellitus and other eye diseases, but also the screening coverage of diabetes mellitus, thus reducing the labor cost of screening.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/application-of-a-scalable-perturbation-algorithm-for-variable-selection-in-uk-biobank-data

Application of a scalable perturbation algorithm for variable selection in UK Biobank data

Last updated:
ID:
79325
Start date:
21 April 2022
Project status:
Current
Principal investigator:
Professor Zhezhen Jin
Lead institution:
Columbia University, United States of America

We aim to (1) identify genetic loci that are associated to quantitative health-related measurements (e.g. BMI, cardiovascular traits, dementia score) (2) identify the relationship between physical activity and health-related outcomes (e.g. type 2 diabetes, cancer) using Biobank data and our proposed algorithm for large datasets. Large datasets such as UK Biobank dataset is ideal for analysis these associations; however, conventional statistical methods lack the scalability to handle processing of such large sample size because the full sample may well exceed the physical memory of an ordinary computer. We propose a novel algorithm based on subsampling to generate a robust and computationally efficient estimator for variable selection and statistical inference in the analysis of big data. The new method will improve our ability to process the full cohort of data with limited computer sources, and the analyses can provide more precise result and better understanding of the association between genes and health status, human activities and health status as well as their underlying biological process. Our project will last for about 3 years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/application-of-ai-algorithms-in-auditory-system-studies

Application of AI Algorithms in Auditory System Studies

Last updated:
ID:
461910
Start date:
13 April 2025
Project status:
Current
Principal investigator:
Mr Xiaotao Liu
Lead institution:
Southeast University, China

Introduction: Hearing impairment is a growing global issue due to aging populations and noise pollution. AI offers innovative solutions for diagnosing and treating hearing impairments and enhancing hearing devices. This research leverages AI to tackle key challenges in auditory science, improving hearing health.

Background: Hearing loss, often caused by noise, medications, genetics, or infections, is traditionally diagnosed through clinical tests that are not always accessible. AI, especially through machine learning (ML) and deep learning (DL), is transforming hearing health by enabling advanced data analysis, diagnostics, and device improvements.

Objectives: This study aims to enhance diagnostic accuracy using ML on hearing test data, optimize hearing devices with DL for better performance in complex environments, and develop smartphone applications for remote hearing health monitoring.

Content: The research includes building ML models to analyze hearing data, applying DL for noise suppression and speech recognition in devices, and integrating AI with apps for remote monitoring and diagnosis.

Methods: Data collection and preprocessing will involve dimensionality reduction and feature selection for model efficiency. The development and testing of ML and DL models will be conducted using Python and TensorFlow, followed by integration into devices and extensive user testing.

Outcomes: The research seeks to create an AI-based diagnostic system, improve hearing device functionality, and establish a remote monitoring platform for broader accessibility to hearing health services.

Conclusion: By utilizing AI, this research aims to significantly advance auditory science, leading to better diagnostics, optimized hearing devices, and increased access to hearing health care globally.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/application-of-artificial-intelligence-ai-in-medical-diagnosis-based-on-liver-imaging

Application of Artificial Intelligence (AI) in Medical Diagnosis Based on Liver Imaging

Last updated:
ID:
62135
Start date:
27 July 2020
Project status:
Closed
Principal investigator:
Mr James Hacunda
Lead institution:
Takeda Pharmaceutical Company Limited, Japan

Fatty liver disease (FLD) is a common disease, with no or very mild symptoms, affecting approximately 1 out of 4 people. Excess fat builds up in the liver, causing health problems. Alcohol consumption may affect the onset or severity of this disease, but it is also prevalent in people that consume little or no alcohol. This version of FLD is referred to as non-alcoholic fatty liver disease (NAFLD).

No medicines are available to treat NAFLD; only changes in diet, alcohol consumption, weight loss, and exercise are recommended by medical professionals.

If NAFLD is not properly managed it can progress to complications. Cirrhosis, a condition where scar tissue builds up damaging the liver is the end stage of NAFLD. When the liver is scarred, high blood pressure in the liver can occur in blood vessels in the esophagus and lead to bleeding issues, or the scarring can also lead to liver cancer, which both can be fatal.

The research planned will create new tools for physicians to perform early diagnosis of liver disease from a non-invasive test, magnetic resonance imaging (MRI), and extending the learnings to the common tool of ultrasound. Usually, doctors detect, characterize, and monitor diseases by assessing pictures of the liver visually, but physicians are human, and are affected by their learning and biases, and may not be able to observe subtle changes. This project will develop innovations using artificial intelligence to recognize liver features and changes in those features automatically with the potential to assist physicians in making more accurate and reproducible imaging diagnosis significantly reducing a physicians’ workload. Because fatty liver is so common, we believe that making a measurement tool that can be used with multiple common pieces of equipment (MRI and Ultrasound) will ultimately improve the public’s health and enable new medicines to be discovered and developed.

The research project to develop the automated imaging characterization tools for researchers and physicians to use in assessing FLD is anticipated to take 24 to 36 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/application-of-artificial-intelligence-ai-in-the-diagnosis-and-prognosis-of-alzheimers-disease

Application of Artificial Intelligence (AI) in the Diagnosis and Prognosis of Alzheimer’s Disease

Last updated:
ID:
749021
Start date:
23 May 2025
Project status:
Current
Principal investigator:
Dr Ha Thi Thanh Huong
Lead institution:
International University, Vietnam National University of Ho Chi Minh City, Viet Nam

Research Questions:
1. Can Alzheimer’s disease (AD) be reliably detected through ocular biomarkers (Fundus and OCT imaging) with comparable accuracy to traditional neuroimaging methods?
2. What are the optimal AI architectures and preprocessing methods for analyzing Vietnamese population-specific imaging data for AD detection?
3. How does the integration of multimodal data (MRI, OCT, Fundus, and clinical data) improve the performance of AI-based AD diagnosis compared to single-modality approaches in the Vietnamese population?
4. How effectively can AI models predict AD progression at 12 and 24 months based on baseline imaging and clinical data, and which biomarkers best correlate with cognitive decline?
Objectives:
1. To develop a comprehensive clinical dataset, including Fundus images, OCT images, and structural brain MRI from 400 Vietnamese subjects to train and validate AI models.
2. To develop and evaluate AI diagnostic models achieving: !80% accuracy from ocular imaging alone; !85% accuracy from MRI alone; !90% accuracy from combined imaging data; and !93% accuracy when adding clinical data.
3. To develop progression prediction models estimating cognitive decline at 12 and 24 months based on baseline data.
Scientific Rationale: The scientific foundation for this research includes:
1. Established biological connections between retinal and brain pathology in AD, where shared neurovascular pathways and amyloid/tau accumulation manifest in both tissues
2. Documented performance variation when AI models trained on Western populations are applied to Asian cohorts (accuracy dropping from 96.8% to 79.2%)
3. Complementary information from different imaging modalities addressing distinct aspects of AD pathology
4. Ocular imaging advantages in capturing early vascular and neural changes preceding cognitive symptoms
5. Evidence that baseline imaging biomarkers correlate with future cognitive trajectory


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/application-of-artificial-intelligence-for-screening-of-risk-factors-and-prediction-of-disease-progression-for-glaucoma-and-myopia

Application of Artificial Intelligence for Screening of Risk Factors and Prediction of Disease Progression for Glaucoma and Myopia

Last updated:
ID:
95829
Start date:
12 May 2023
Project status:
Current
Principal investigator:
Professor Yehong Zhuo
Lead institution:
Zhongshan Ophthalmic Center, China

1.Aim:
1.1. To investigate an artificial intelligence algorithm for glaucoma, myopia, and myopia combined with glaucoma (MG) based on medical images.
1.2. To explore the factors associated with glaucoma, myopia and MG, including patients’ genetic information, systemic diseases, and living environment.

2. Scientific rationale:
2.1 Significance of the research project
Glaucoma is a progressive and potentially blinding eye disease. Fundus photography and optical coherence tomography (OCT) are practical tests for early glaucoma screening. Myopia is a global public problem and can double the risk of glaucoma. The optic disc of myopic eyes may appear tilted or stretched on the fundus photograph, similar to glaucoma. In OCT, the retinal nerve fiber layers defect potentially leading to a misdiagnosis of glaucoma. Therefore, developing more effective screening approaches for myopia and glaucoma are valuable.
2.2 Research status
Besides the traditional fundus examinations, multimodal brain fMRI provides a new understanding of glaucoma and myopia. Further correlation analysis between the eye and brain function may help the differential diagnosis.
AI has been applied in detecting glaucoma and myopia, and the researchers have shown its application prospect. However, little progress has been made in differentiating myopia from glaucoma. Here we attempt to develop a medical image algorithm and explore the influencing factors for glaucoma and myopia based on AI.

3. Project duration
The project will last for 3 years

4. Public health impact
4.1. Considering a wide range of examinations, our AI algorithm for screening glaucoma, myopia, and MG could assist in precise diagnosis and efficient therapy.
4.2. Our study will establish a comprehensive model including genetic and environmental information. The research will provide new diagnostic ideas and early prevention.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/application-of-artificial-intelligence-in-tumor-diagnosis-and-treatment

Application of artificial intelligence in tumor diagnosis and treatment

Last updated:
ID:
98680
Start date:
12 November 2024
Project status:
Current
Principal investigator:
Professor Xinwei Han
Lead institution:
First Affiliated Hospital of Zhengzhou University, China

Aims: we aim to provide a stable and widely applicable method for tumor diagnosis and treatment based on clinical big data combined with Artificial intelligence methods.

Scientific rationale: Cancer is one of the leading causes of death worldwide, with 19.29 million new cancer cases in 2020, down 18.3% from a year earlier; in 2021, there were more than 20 million new cases of cancer worldwide. Cancer patients carry a heavy disease burden and have huge unmet needs. Artificial intelligence (AI) is important for risk prediction, early detection of diseases, diagnosis through sequencing and medical imaging, accurate prognosis, biomarker identification, and therapeutic target identification for new drug discovery. To this end, we analyzed and summarized the application of different artificial intelligence methods in tumor diagnosis and treatment, hoping to provide a stable and widely applicable method for tumor diagnosis and treatment based on clinical big data combined with AI methods. The study aims to demonstrate the feasibility of a new integrated AI diagnosis and treatment system and apply it in clinical practice.

Project duration: Three years will be needed to complete this project.

Public health impact: This project is expected to significantly improve cancer risk prediction and prevention, enhance the accuracy of cancer screening and diagnosis, better stratify cancer risk, and guide us to more accurately identify high-risk individuals, thereby helping cancer patients provide more precise treatment strategies. This research will make an important contribution to the prognosis of cancer patients and greatly reduce the burden of cancer patients.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/application-of-deep-learning-for-investigating-various-biomarkers-related-to-retinal-images

Application of deep learning for investigating various biomarkers related to retinal images

Last updated:
ID:
179578
Start date:
15 April 2024
Project status:
Current
Principal investigator:
Professor Daniel Duck-Jin Hwang
Lead institution:
Lux Mind., Korea (South)

Aims: The primary objective of this research is to utilize fundus photography and OCT scan data to analyze and predict prospective or existing cardiovascular, cerebrovascular, or renal diseases. The central focus is developing a deep learning model based on retinal images, which will contribute to the anticipation of cardiovascular, cerebrovascular, or renal diseases risks and aid in the early diagnosis and prevention of these disorders. This study aims to harness the potential of retinal imaging and advanced deep-learning techniques to enhance our understanding of heart, brain, kidney health and improve clinical outcomes through proactive interventions.

Scientific raionale: This study investigates the application of deep learning technology through retinal imaging to examine the correlation between cardiovascular, cerebrovascular, or renal diseases and retinal status. Previous research has raised the hypothesis of a link between the retina and these non-ophthalmic diseases, highlighting the potential for using retinal images in predicting theses diseases. However, more studies are needed to explore this association comprehensively. This research endeavors to bridge this scientific gap by developing highly automated models and assessing their practicality in clinical settings. It seeks to advance our understanding of the relationship between retinal health and theses disorders while exploring the clinical applications of this innovative approach.

Project duration: 36 months

Public health impact: The paramount public health impact of this research lies in the early diagnosis and prevention of cardiovascular, cerebrovascular, or renal diseases using retinal images. The developed deep learning model allows for a more precise measurement of a patient’s biological age, facilitating the provision of personalized preventive strategies. This approach is expected to enhance disease early detection and patient management efficiency within the public health system. Furthermore, this research can potentially promote a shift toward a prevention-oriented healthcare system, supporting healthier aging and alleviating the burden of chronic diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/application-of-deep-learning-for-the-characterisation-of-motion-patterns-in-the-heart

Application of deep learning for the characterisation of motion patterns in the heart

Last updated:
ID:
40161
Start date:
5 September 2018
Project status:
Current
Principal investigator:
Professor Vicente Grau
Lead institution:
University of Oxford, Great Britain

The heart requires a complex combination of mechanical, chemical and electrical phenomena to produce and sustain an efficient beating action. Motion of cardiac walls during contraction and subsequent relaxation can be captured by modern imaging methods such as Cardiac Magnetic Resonance. It is known that such motion patterns are affected by disease, but current quantification methods leave substantial room for improvement.
Machine learning, and in particular the more recent methods known as deep learning, has revolutionised several areas of research such as computer vision. Its effective use requires the availability of large data sets, which had been lacking until the appearance of large data collection initiatives such as UK Biobank.
The proposed research aims at developing deep learning methods for the analysis of motion patterns in hearts from the UK Biobank, learning models of normal motion as well as the natural variability in the Biobank cohort, establishing links between these models and different subject characteristics, and eventually allowing the prediction of cardiac risks.
The proposed research plan will be carried out initially over a 36 month period, with the expectation that further research questions are developed over that time leading to subsequent scientific questions and potential new projects.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/application-of-deep-learning-systems-for-detecting-glaucomatous-optic-neuropathy

Application of deep learning systems for detecting glaucomatous optic neuropathy

Last updated:
ID:
55836
Start date:
2 November 2020
Project status:
Current
Principal investigator:
Ms Zhixi Li
Lead institution:
Sun Yat-Sen University, China

Glaucoma is one of the leading causes of irreversible vision loss and blindness worldwide. Early screening and diagnosis of glaucoma is made difficult due to the asymptomatic nature of the disease in its early stages and subjective approaches to diagnosis. Recent evidence has indicated that deep learning based artificial intelligence screening systems which have been applied to the detection of ophthalmic diseases can achieve excellent sensitivities and specificities, suggesting a revolutionary improvement in disease screening. The purpose of our research is to further evaluate the performance of a deep learning system for the detection of referable glaucomatous optic neuropathy in different population settings.

Using fundus and OCT images as the primary inputs, we will further validate the accuracy of a deep learning algorithm to identify referable glaucomatous optic neuropathy, which has previously been developed and validated basing on 48,116 fundus images from different clinical settings in China.

Based on the above, we are requesting data from all patients (68,151) that have had retinal imaging performed in UK Biobank. The validation of our deep learning algorithm will be based on medical information such as age, gender and diagnoses, and results from eye measurements, such as intraocular pressure (IOP), fundus photographs and OCT.

If the efficacy of this deep learning algorithm is successfully proven, this work will help to improve the detection of glaucoma and potentially other ophthalmic diseases. Other vital potential benefits include improving screening coverage and reducing healthcare costs due to earlier diagnosis.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/application-of-deep-machine-learning-to-predict-cardiovascular-outcomes

Application of deep machine learning to predict cardiovascular outcomes.

Last updated:
ID:
54050
Start date:
23 December 2019
Project status:
Current
Principal investigator:
Professor Taane G Clark
Lead institution:
London School of Hygiene and Tropical Medicine, Great Britain

Asymptomatic left ventricular dysfunction (ALVD) of the heart is present in up to 6% of the UK population. ALVD is associated with reduced quality of life and longevity, and is treatable when diagnosed. An inexpensive, noninvasive screening tool for ALVD would assist clinical making. An electrocardiogram (ECG) is a routine test to measure the heart’s electrical activity. By applying a machine learning approach to ECG data from the UK Biobank it will be possible to develop a model that predicts ALVD, as well as participant characteristics such as age and gender. The performance of the model will we assessed on ECG data from other populations. Ultimately, the validation of such approaches to predict cardiovascular outcomes could assist clinical decision making and public health initiatives. The duration of the project is twelve months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/application-of-deep-neural-networks-to-human-ageing-biomarker-development

Application of deep neural networks to human ageing biomarker development

Last updated:
ID:
41714
Start date:
14 September 2018
Project status:
Closed
Principal investigator:
Dr Qingsong Zhu
Lead institution:
Insilico Medicine, Inc, United States of America

Ageing is a complex process that has been observed in all biological systems at every level of organisation. Some anti-ageing interventions have been demonstrated life-extending effects in model organisms. However, the translation of these interventions in human clinical practice remains limited. The absence of comprehensive ageing biomarkers is one of the major impediments for the translation to the clinic. At the same time, multiple biomarkers proposed today are expensive and not as practically measurable (lack of the standardize assays and etc). The most accurate methods of calculating biological age are a subject of ongoing debate, and recent studies suggest that a suite of biomarkers, rather than any individual biomarker, constitute the most effective means of assessing the health status. It has also been shown that ageing biomarkers are population-specific, hence the age predicting models are needed to be trained upon population-specific data (Mamoshina et al., 2018). In this project, we aim to train the deep learning model (Deep Neural Networks) using UK Biobank datasets to validate the previously discovered biomarkers and to complete and complement the robust ageing biomarkers that may be easily targeted and measured. The developed biological age predicting measures will be based on the data, that most of the people already have, such as blood test and urine test, and electrocardiograms, and passive recordings, such as wearable devices. The results of the current research will lead to simple, minimally invasive, and affordable methods of tracking integrated biomarkers of ageing in humans.

Mamoshina P, Kochetov K, Putin E, Cortese F, Aliper A, Lee WS, Ahn SM, Uhn L, Skjodt N, Kovalchuk O, Scheibye-Knudsen M, Zhavoronkov A / Population specific biomarkers of human aging: a big data study using South Korean, Canadian and Eastern European patient populations // J Gerontol A Biol Sci Med Sci. 2018


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/application-of-fast-mixed-model-association-and-principal-component-analysis-methods

Application of fast mixed model association and principal component analysis methods

Last updated:
ID:
10438
Start date:
1 May 2015
Project status:
Closed
Principal investigator:
Dr Alkes Price
Lead institution:
Harvard School of Public Health, United States of America

We aim to identify genetic loci that are associated to specific health-related outcomes. More precisely, we will apply a new, more powerful statistical method (BOLT-LMM) to analyze outcomes that have been demonstrated to be heritable in previous genome-wide association studies, including direct health outcomes (disease status) as well as heritable quantitative measurements such as height, BMI and lipid levels associated to some health outcomes. We will investigate only genetic effects and will use environmental exposure data only as covariates in our analyses. This project is restricted to self-reported outcomes and traits measured at baseline. Our discovery of associated loci that could not be discovered using existing methods may potentially lead to actionable drug targets, and is in the public interest. We will analyze each outcome independently: i.e., for each disease code, we will compute association statistics between all genetic markers and the disease code (independent of other outcomes). We will apply a more powerful statistical method to the data than has previously been available. The new method (BOLT-LMM) applies a linear mixed model to analyze all genetic markers simultaneously, enabling a more powerful statistical analysis that is expected to detect associations that other methods miss. In addition to performing BOLT-LMM analysis, we will also compute association statistics using other standard methods for comparison. We will analyze the full cohort.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/application-of-fine-grained-aggregation-for-rare-variant-analysis-with-biobank-scale-cohorts

Application of Fine-Grained Aggregation for Rare Variant Analysis with Biobank-scale Cohorts

Last updated:
ID:
266889
Start date:
25 October 2024
Project status:
Current
Principal investigator:
Dr Robert Altshuler
Lead institution:
AIRNA, Inc., United States of America

A fundamental challenge in developing new therapies for human diseases is identifying the genes and genetic variants that are involved and the biological mechanisms through which they cause diseases, including whether the diseases result from increases or decreases in the functional activity of the genes. This is especially difficult for many common diseases, such as heart disease, because hundreds of genes can contribute to disease risk. Although rare variants are believed to have an important role in causing common diseases, historically, this has been difficult to investigate. Recently, as the costs of genome sequencing have decreased, and biobanks have been established that contain medical records and genetic data for hundreds of thousands of participants, it has become more feasible to study this topic.
The past few years have seen the publication of multiple research studies using data from hundreds of thousands of biobank participants to examine the role of rare variants in disease. By design, these studies have been expansive, considering rare variants in tens of thousands of genes and their combined effects on thousands or tens of thousands of diseases or related traits. The massive scale of these studies allowed them to produce an incredible volume of results, but also came with limits on the statistical power with which they could detect the associations of rare variants with disease, which made it challenging to translate the results into new therapies.
In this project, which we expect will go on for 24 months, in addition to using latest and largest dataset of rare variants for all of the UK Biobank participants, we’ll apply a very targeted approach, focusing on a small number of common diseases and related clinical measurements, for example heart disease and LDL cholesterol. Furthermore, testing for associations with rare variants in a limited set of genes that are known to be relevant for the selected diseases, will enable a much more fine-grained approach than in previous studies for how the variants and their effects are combined into groups. All together, this project will have increased power to detect rare variants associated with disease and will produce results that can point more directly to the mechanisms through which disease is caused. In particular, the project’s fine-grained approach will help identify the specific parts of proteins where changes affect disease risk, and make it easier to discover variants that have protective effects, facilitating the development of new therapies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/application-of-genetic-environment-and-lifestyle-data-to-understand-causal-risk-factors-of-complex-metabolic-alterations-and-develop-novel-prediction-model

Application of genetic, environment and lifestyle data to understand causal risk factors of complex metabolic alterations and develop novel prediction model

Last updated:
ID:
70522
Start date:
7 February 2022
Project status:
Current
Principal investigator:
Dr Chuncheng Lu
Lead institution:
Nanjing Medical University, China

Background: The metabolic dysfunctions can result in the development of various associated metabolic diseases, including type 2 diabetes mellitus (T2DM), cardiovascular disease (CVD), nonalcoholic fatty liver disease (NAFLD) and cancer. Genetic, environmental and lifestyle factors are the main risk factors on metabolic syndrome. However, the causal effects of the genetic, environmental or lifestyle factors and metabolic alterations remain to be elucidated.
Objective: This study aims to explore the causal associations between genetic, lifestyle or environmental factors and the risk of metabolic diseases using Mendelian randomization (MR) analysis.
Project duration: The duration of this project will be three years, from 2021-2023.
Public health impact: This research will provide important contributions to advance understanding of environmental, lifestyle or genomic influence and mechanisms underlying complex metabolic traits. We will finally build novel strategies for risk prediction in the common metabolic diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/application-of-genome-wide-association-study-gwas-in-drug-repositioning-taking-ankylosing-spondylitis-as-an-example

Application of Genome Wide Association Study (GWAS) in Drug Repositioning -Taking Ankylosing Spondylitis as an Example

Last updated:
ID:
67903
Start date:
15 February 2021
Project status:
Current
Principal investigator:
Miss Mengting Lee
Lead institution:
National Defense Medical Center, Taiwan, Province of China

Ankylosing spondylitis (AS) is an autoimmune disease. In addition, genetics have been considered the main cause of the disease. Since there is still no cure for AS, only drugs, exercises, and surgery can be used to delay the worsening of symptoms.

Drug repositioning refers to the search for compounds that are already on the market or in the advanced clinical stage. The indication may be other diseases, but it may also be used to treat other new indications.

We expect to spend 36 months exploring the difference between the susceptibility gene of AS between the Taiwanese ethnic group and European whites and screening out new potential drug candidates for treatment from existing or research drugs.

Based on the results of genetic testing, clinicians can give patients medication recommendations that better meet individual needs.
Repositioning drugs to find potential drugs that can treat ankylosing spondylitis from existing drugs can provide new treatment options.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/application-of-imiomics-to-whole-body-mri-data-for-creation-and-application-of-a-human-imaging-atlas

Application of Imiomics to whole-body MRI data for creation and application of a human imaging atlas

Last updated:
ID:
14237
Start date:
10 September 2018
Project status:
Current
Principal investigator:
Professor Joel Kullberg
Lead institution:
Uppsala University, Sweden

We are facing a global epidemic of obesity and related cardiovascular complications. This calls for novel intervention strategies where improved understanding of the underlying mechanisms is key. Both total fat mass and its distribution throughout the body have been linked to development of cardiovascular disease.

Whole-body medical imaging using magnetic resonance (MRI) can sample anatomical information such as tissue volume and fat content, i.e. parameters relevant for cardiovascular studies, at the millimetre scale using millions of small 3D-elements (voxels). We have developed and validated a novel technology, Imiomics, which enables whole body voxel-wise correlations with non-imaging data, e.g. genotypes and disease phenotypes. Imiomics thereby allows innovative types of whole body composition studies. During the Imiomics analysis, whole-body images are registered/deformed to a common coordinate system/geometry. This allows statistical analysis, in the whole-body region, such as creation of a `mean person` (atlas) and studies of deviations from that mean person. This also allows integration of imaging and non-imaging data as whole-body `correlation- images` to for example blood parameters can be made.

The aim of this project is to determine causes and consequences of variation in human body composition. This goal will be achieved by
1) applying Imiomics to build a Human Imaging Atlas using whole-body imaging data from large-scale cohort studies
2) genome-wide body-wide studies of body composition
3) assessment of the association of body composition with cardiovascular disease, including ischemic stroke and myocardial infarction and cardiovascular risk factors, including type 2 diabetes, hypertension, and dyslipidemia
4) Mendelian Randomization studies of causal effects.

This interdisciplinary project addresses several fundamental questions related to causes and consequences of variation in body composition. This will improve our understanding of the underlying mechanisms of cardiovascular disorders and accelerate development of prevention, diagnosis and treatments for cardiovascular complications. Furthermore, the resources created by this project are anticipated to open up new avenues for research within the obesity-field.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/application-of-machine-learning-and-risk-prediction-model-in-late-life-diseases

Application of machine learning and risk prediction model in late-life diseases

Last updated:
ID:
94653
Start date:
14 December 2022
Project status:
Current
Principal investigator:
Dr Xi-jian Dai
Lead institution:
Nanchang University, China

This project will find what and where our brain changed when we have a diagnosis of late-life disease. These finding may broaden and expand our understanding of the neurobiological mechanism of late-life diseases.
Furthermore, we will find risk and protective factors for late-life diseases. Next, we will develop a risk prediction system based on these factors to tell us what is the risk of developing this disease in the future. This system will could help us to identify our potential risk profile and provide guidance on precise and timely actions to prevent or delay to develop late-life disease.
In view of huge works on data cleansing and data analyses, this project will last for 36 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/application-of-machine-learning-for-classification-of-late-life-diseases-and-their-outcome-prediction

Application of machine learning for classification of late-life diseases and their outcome prediction

Last updated:
ID:
75732
Start date:
24 February 2022
Project status:
Current
Principal investigator:
Professor Yongjun Wang
Lead institution:
Shenzhen Kangning Hospital, China

First, we use the machine learning algorithms to differentiate patients with late-life diseases from those without late-life diseases for early diagnosis, and tell us what and where our brain changed.
Second, we used the magnetic Resonance Imaging (MRI) data at baseline to predict their outcome endpoints of late-life diseases in the longitudinal data at follow-up, and tell us what and where our brain changed when one disease progress to another disease.
Third, we used the MRI data at baseline to predict their brain age, and tell us whether and why our brain are elder when we have a diagnosis of late-life diseases.
Fourth, we will use the baseline data to build a risk prediction and evaluation system. This system will tell you the risk of developing a late-life disease when you have poor lifestyle or other adverse events.
These finding may broaden and expand our understanding of the neurobiological mechanism of late-life diseases. In view of huge works on data cleansing and data analyses, this project will last for 36 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/application-of-machine-learning-for-depression-classification-and-outcome-prediction

Application of machine learning for depression classification and outcome prediction

Last updated:
ID:
58228
Start date:
5 May 2020
Project status:
Closed
Principal investigator:
Dr Jihui Zhang
Lead institution:
Chinese University of Hong Kong, China

Currently, the diagnosis of depression is mainly based on clinical presentations and there is a lack of reliable markers to differentiate patients with depression from those without depression. Furthermore, the neurobiological mechanism of depression has been rarely understood. The machine learning approach can train a classifier for early diagnosis and outcome prediction, and propose a network hypothesis for disease. In this project, we applied the machine learning algorithms into depression for this purpose.
First, we use these algorithms to differentiate patients with depression from those without depression for early diagnosis; Second, we used the MRI data at baseline to predict the outcome endpoints of the depression in the longitudinal data at follow-up; Third, we will identify whether patients with depression have hippocampal volume decrease because this area is very important for cognitive function. Third, the Mendelian randomization analysis will be used to investigate the possible causal relationship among the genes, hippocampal volume, brain functional alterations, and various outcomes (depression score, anxiety score, sleep parameters).
These findings could help us early diagnosis and outcome prediction for the depression, and tell us whether depression subjects have hippocampal atrophy, and their relationships with genes, sleep, depression symptom or other variables. Furthermore, these finding may broaden and expand our understanding of the neurobiological mechanism of depression. In view of huge works on data cleansing and data analyses, this project will last for 36 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/application-of-machine-learning-methodologies-on-the-uk-biobank-dataset-to-predict-onset-of-depression

Application of machine learning methodologies on the UK biobank dataset to predict onset of depression

Last updated:
ID:
83122
Start date:
12 April 2022
Project status:
Current
Principal investigator:
Yonatan Bilu
Lead institution:
K I Research Institute (R.A), Israel

We aim to identify middle-aged adults who are at risk for future depression. Depression is a major cause of disability globally, with a peak of prevalence among middle-aged adults. Identifying a combination of factors that are most predictive of upcoming depressive episode in middle-aged individuals is crucial for developing prevention strategies. Previous work have shown the great potential of mahcine-learning techniques for similar goals. We expect that the analysis can be done in 6 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/application-of-machine-learning-methodologies-on-the-uk-biobank-dataset-to-quantify-the-effect-of-lifestyle-habits-on-health

Application of machine learning methodologies on the UK biobank dataset to quantify the effect of lifestyle habits on health

Last updated:
ID:
58770
Start date:
27 March 2020
Project status:
Current
Principal investigator:
Dr Pinchas Akiva
Lead institution:
K I Research Institute (R.A), Israel

The primary aim of this project is to utilize the large data set available in the UK biobank to establish a quantitative association between the lifestyle behavior (e.g. diet, fitness activity, smoking habits, stress, etc.) of the subjects and their health status. The association between specific lifestyle behaviors and chronic diseases is widely studied (e.g. sedentary lifestyle and cardiovascular disease risk, consumption of processed meat and cancer, etc.). However, quantifying such risks according to lifestyle behavior remains a challenge, as well as measuring the combined effect of multiple behaviors on multiple outcomes. By analyzing data of the entire UK Biobank cohort, including lifestyle questionnaire data, primary care records, and additional clinical data records, we will construct multiple machine learning models tailored to predicting a wide set of disease risks. In addition, we plan to study the association of lifestyle behaviors to diseases through clinical biomarkers (e.g. CRP, ESR, glucose, albumin, BMI, TG, cholesterol, BP, etc.). This will allow us to study how lifestyle behavior affects the clinical biomarkers, and how these biomarkers can be used for health status evaluation (i.e. how clinical biomarkers mediate the association between lifestyle behavior and clinical status). We will undertake this research using anonymous data. The project will last 18 months.
The high impact of this study stems from its ability to influence a very wide population. An ability to better understand and quantify the effect of lifestyle behavior on health has the potential to allow choosing the most fitting change, considering the personalized impact on the individual’s health. This project will potentially expand the public knowledge concerning lifestyle habits and their impact on disease risk, and will also contribute to the creation of models that will better allow assessing and improving health, thus avoiding major health complications in the future.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/application-of-multi-omic-predictors-for-predixcan-analysis-and-functional-genetic-risk-prediction-to-lung-function-and-copd-in-the-uk-biobank

Application of multi-omic predictors for PrediXcan analysis and functional genetic risk prediction to lung function and COPD in the UK Biobank

Last updated:
ID:
59234
Start date:
25 January 2021
Project status:
Closed
Principal investigator:
Dr Ani Manichaikul
Lead institution:
University of Virginia, United States of America

Chronic lower respiratory disease (CLRD), which includes emphysema and COPD (chronic obstructive pulmonary disease), is a leading cause of morbidity. COPD is diagnosed by a decrease in lung function, namely forced expiratory volume in one second (FEV1) and its ratio to forced vital capacity. Deeper understanding of the underlying biology and identification of drug targets are urgently needed. Several genetic risk factors have been identified but further research is needed to find actionable targets. To address this need, we will use an integrative analysis framework to identify genes and proteins associated with lung function and COPD in the UK Biobank. We will further combine the insights gained from our analyses to build genetic risk prediction models . We expect our resulting prediction models will be more biologically interpretable than many existing genetic risk prediction models.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/application-of-multi-omics-analysis-based-on-artificial-intelligence-in-diagnosis-and-treatment-of-cognitive-disorders-and-neurodegenerative-diseases

Application of multi-omics analysis based on artificial intelligence in diagnosis and treatment of Cognitive Disorders and Neurodegenerative Diseases.

Last updated:
ID:
102865
Start date:
27 November 2024
Project status:
Current
Principal investigator:
Professor Jun Liu
Lead institution:
Guangzhou Medical University, China

The research focus on exploring the underlying mechanisms and disease development processes of cognitive disorders and neurodegenerative diseases such as Alzheimer’s disease and Parkinson’s disease. By combining the living habits, clinical information, biological information and imaging information of patients in the database, we integrate the scientific system of the occurrence and development of these diseases. Our work has been well underway for two years and will continue for a long time. More importantly, we hope to build a diagnostic system for early screening of cognitive disorders and a prediction model for the diagnosis and treatment of neurodegenerative diseases. In addition, more early risk factors for disease will be identified, so as to provide early, timely and effective prevention guidance for the public.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/application-of-multi-omics-analysis-in-the-risk-complications-biomarker-treatment-sensitivity-and-pathogenesis-of-insomnia

Application of multi-omics analysis in the risk, complications, biomarker, treatment sensitivity, and pathogenesis of insomnia.

Last updated:
ID:
171240
Start date:
8 October 2024
Project status:
Current
Principal investigator:
Dr Qianzi Yang
Lead institution:
Ruijin Hospital, China

Aim: Our research aims to establish an assessment model for insomnia risk factors and identify biomarkers associated with insomnia and its complications.
Scientific rationale: Through deep learning, a risk prediction model can be established to identify high-risk factors for insomnia. Bioinformatics can process and analyze large-scale data. For example, methods such as gene expression profiling, pathway analysis, transcription factor recognition, and protein-protein interaction network analysis can identify potential biomarkers related to insomnia.
project duration: A year
public health impact: The identification of high-risk factors for insomnia will enable early intervention and promote public health education. Furthermore, the biomarkers of insomnia and its complications, as well as the features observed in neuroimaging, will enhance our understanding of the mechanisms underlying insomnia and guide clinical practice.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/application-of-polygenic-methods-to-investigate-shared-and-unique-genetics-across-psychiatric-traits-including-bipolar-disorder-depression-and-substance-use-disorders

Application of polygenic methods to investigate shared and unique genetics across psychiatric traits including bipolar disorder, depression and substance use disorders

Last updated:
ID:
55108
Start date:
6 January 2020
Project status:
Current
Principal investigator:
Professor Joanna Biernacka
Lead institution:
Mayo Clinic, United States of America

Research has shown that psychiatric traits are influenced by many genetic variations with very small individual effects (i.e. these traits are highly polygenic), and statistical methods that account for the effects of many genetic variations (i.e. polygenic methods) have become widely used in psychiatric genetics research. Polygenic methods can help identify shared genetic risk factors across different disorders. For example, it has been shown that bipolar disorder shares genetic risk factors with schizophrenia. An understanding of the shared genetics across disorders can also help reveal genetic heterogeneity within disorders (e.g. schizophrenia genetic risk factors are shared especially with bipolar disorder with psychosis, and less so with bipolar disorder without psychosis). A better understanding of such shared genetics is improving our ability to identify the genetic contributors to these complex traits.
As a statistical genetics research group dedicated to understanding the genetic underpinnings of psychiatric traits and individual differences in response to their treatment, we use polygenic methods to study genetic heterogeneity that gives rise to disease subtypes, which can be defined by symptoms or comorbidities. To make further progress in characterizing the genetic heterogeneity of complex psychiatric diseases, we are extending currently used statistical methods to improve subtype prediction from genetic data. These methods need to be evaluated and applied in large datasets, and the UK Biobank will be an ideal dataset to advance this work. Specifically, we will:
1. Develop new analytical approaches, including extensions of polygenic risk score and machine learning methods, including new strategies for modeling gene-environment interactions, to improve genetic prediction of psychiatric traits. UK Biobank data will be used to evaluate the new methods.
2. Develop predictive models for psychiatric disorders, focusing on bipolar disorder and substance use disorder, and their intersection. We will consider predictive models that incorporate polygenic risk scores for multiple related traits and risk factor interactions, and will compare to prediction based on machine learning approaches. We will also use genetic data to model effects of possible intermediate risk factors such as gene expression and metabolite levels that may be altered in patients with psychiatric disorders, and incorporate these effects in psychiatric disorder prediction.
We expect our research to provide insights into genetic risk for psychiatric traits, which may ultimately improve clinical risk assessment and prediction of prognosis and treatment response. We also expect to develop statistical method that may prove useful in detecting genetic risk factors for other medical conditions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/application-to-contact-patients-regarding-treatment-in-morning-vs-evening-time-study

Application to Contact Patients regarding Treatment In Morning vs Evening (TIME) Study

Last updated:
ID:
12243
Start date:
1 September 2015
Project status:
Closed
Principal investigator:
Professor Thomas MacDonald
Lead institution:
University of Dundee, Great Britain

To determine if taking once a day anti-hypertensive medication is more efficacious taken in the evening compared with the morning. If a change to evening dosing is more efficacious than usual morning dosing and is acceptably safe then this would be of major benefit to public health. Hypertension is widespread throughout the population and thus if TIME can help establish if there is an optimum time for anti-hypertensive dosing then this will have a significant impact on improving patient care in this area across the whole of the UK. A pilot-phase has recruited ~400 participants via general-practices, using letters, emails and posters (about 1 per 30 invitations). We would like to boost recruitment to the study through inviting UKB participants to join if they fulfil the eligibility criteria (those who currently take blood pressure tablets once a day). We suggest inviting UKB participants who have self-reported hypertension at baseline. These invitations would be sent by email. If this insufficient we may invite further participants by letter. About 10,000 patients are required to ensure the study is powered sufficiently. The power calculations undertaken to reach this figure are detailed in section 12 of the TIME Study protocol. If the study recruits more participants than this target then follow up time should be reduced allowing the study to report earlier, which would be advantageous. We hope that the UKB resource could help us to achieve this outcome.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/application-to-use-uk-biobank-data-as-part-of-the-psychiatric-genomics-consortium-for-ptsd-neuroimaging-project

Application to Use UK Biobank Data as Part of the Psychiatric Genomics Consortium for PTSD Neuroimaging Project

Last updated:
ID:
40249
Start date:
2 November 2018
Project status:
Closed
Principal investigator:
Dr Rajendra Morey
Lead institution:
Duke University, United States of America

Exposure to trauma and abuse during childhood is a major risk factor for developing psychiatric disorders in adulthood such as post-traumatic stress disorder (PTSD). However, not all children exposed to trauma will develop such disorders. The fact that some people develop PTSD and others do not can be partially accounted for by their genetics; the chance of inheriting a genetic susceptibility to PTSD is approximately 40%. A few recently discovered genes interact with childhood trauma to increase rates of anxiety and mood disorders in adulthood. This risk may be detected in part by looking at brain measures; this is due to the fact that brain measures have simpler underlying genetic than the many factors that play into developing psychiatric disorders.
Our goal is to conduct a genetic analysis of relevant brain measures, with the long-term goal of identifying genes that lead to brain structure variations that are helpful for early prediction and treatment of several psychiatric disorders where childhood trauma is a major risk factor. We hypothesize that (1) childhood trauma will interact with specific genetic markers to produce changes in brain structure and adult psychopathology, and (2) that unique genetic variants, in the context of genetic vulnerability to childhood trauma, will influence the onset of specific disorders (e.g. depression vs PTSD).
While this project will take place over several years, this extended time period allows us to combine data across many different sites and build an unparalleled data set. Over the course of this project we will be able to greatly increase our understanding of why some people develop PTSD as well as provide new guidelines for early diagnosis and treatment of PTSD.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/applying-80-20-rule-to-efficiently-capture-proteome-data

Applying 80-20 rule to efficiently capture proteome data

Last updated:
ID:
410432
Start date:
4 November 2024
Project status:
Current
Principal investigator:
Dr Nathaniel Robichaud
Lead institution:
Nomic Bio Inc., Canada

Protein measurements provide invaluable information to the healthcare system, providing biomarkers that help to diagnose diseases, determine which patients are likely to benefit from a treatment, and tracking how well patients are responding. Discovering these critical biomarkers requires technologies that can initially measure thousands of proteins simultaneously, in order to find the few that matter for a particular test. While measuring thousands of proteins is technically possible, it is extraordinarily expensive; as a results, studies to identify these crucial biomarkers are limited by cost. Of note, there is a wealth of proteomics information in the UK databank, that can help identify the proteins that are most commonly present in the blood and most often associated with an individual’s health. We propose to apply the 80/20 rule to identify the 20% of proteins that capture 80% of biological information. Using this shortlist will enable future biomarker studies to dramatically reduce costs, therefore enabling more studies and the discovery of more biomarkers to improve the diagnosis and treatment of human diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/applying-a-multi-omics-approach-to-investigate-the-persistent-smoking-effect-on-cancer-risk-and-other-smoking-related-diseases-among-former-smokers-and-to-reveal-underlying-biological-mechanisms

Applying a multi-omics approach to investigate the persistent smoking effect on cancer risk and other smoking-related diseases among former smokers and to reveal underlying biological mechanisms

Last updated:
ID:
858244
Start date:
27 August 2025
Project status:
Current
Principal investigator:
Dr Duc Huy Le
Lead institution:
Vanderbilt University Medical Center, United States of America

-Scientific rationale:
Smoking is one of the strongest and most modifiable health risk factors, accounting for an estimated 2.01 million cancer-related deaths (95% UI: 1.68 to 2.4) globally in 2021. Although smoking cessation significantly reduces the cancer and other health risks, elevated health risks, particularly lung cancer risk among former smokers, can persist for many years after quitting smoking. Recent evidence has provided evidence in identifying markers influenced by tobacco smoke exposure and their association with various chronic diseases. However, few studies have integrated multi-omics data (eg, metabolomics, proteomics, and methylation) to reveal the biological mechanisms for disease risk among former smokers. Therefore, we propose the project to address the following research questions.
1. What metabolites, proteomics, and methylation are associated with smoking status and which differences persist among former smokers?
2. Are multi-omics biomarker signatures built to capture the persistent smoking effect associated with the risk of smoking-related diseases (including cancer, diabetes, and cardiovascular diseases) among former smokers?
Objectives of the study are:
1. To develop and validate persistent smoking effect signatures based on existing multi-omics data, including metabolomics, proteomics, and methylation from the UK Biobank.
2. To assess the associations of the residual smoking effect signatures with cancers, diabetes, and CVD risks among former smokers in the UK Biobank.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/applying-artificial-intelligence-to-define-clinical-trajectories-for-personalized-prediction-and-early-detection-of-comorbidity-and-multimorbidity-patterns

Applying ARtificial Intelligence to Define clinical trajectorieS for personalized predicTiOn and early deTEctiOn of comorbidiTy and muLtimorbidiTy pattErnS

Last updated:
ID:
231240
Start date:
18 April 2025
Project status:
Current
Principal investigator:
Dr Rui Providencia
Lead institution:
University College London, Great Britain

Cardiovascular disease (CVD) is a group of common disorders affecting the heart and blood vessels and are among the top causes of death worldwide. According to WHO, they account for up to 30% of all deaths, of which 85% are caused by heart attacks or strokes. These common and acute events are often linked to other underlying problems in the heart or vessels. Heart rhythm disturbances such as atrial fibrillation (AF) is the most common heart arrhythmia and research suggest it is linked to ~20% of all strokes. In European countries management of AF accounts for up to 3% of the total health care expenses, and it is a growing problem predicted to affect 18 million people by 2050.
Doctors have improved predicting of what type of patient that might develop serious problems from CVD, such as strokes or heart attacks, and now have better medicines to prevent serious events. There is, however, still a lot of work to do. Some patients still have strokes when taking these medicines, and others face different issues such as bleeding or heart failure.
If we can improve our ability to predict who is at risk for certain problems caused by CVD, and which treatment will offer the best course for each individual patient, we can make a big difference in how long and how well they live.
Our goal is to improve the clinical prognosis and quality of life for people with CVD by finding new ways to detect patients at higher risk and selecting the right treatments. We aim to achieve this by using the medical records, genetic data, and images from patients in the UK-Biobank, to develop an artificial intelligence tool relying on a mix of well-known and more sophisticated mathematical and computer algorithm strategies. This tool will help us better manage and care for patients with CVD.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/applying-deep-learning-models-for-automated-quantification-of-glaucomatous-damage-from-color-fundus-photographs

Applying deep learning models for automated quantification of glaucomatous damage from color fundus photographs

Last updated:
ID:
611072
Start date:
30 July 2025
Project status:
Current
Principal investigator:
Professor Leopold Schmetterer
Lead institution:
Nanyang Technological University, Singapore

Research Question:
How effectively can a trained deep learning model (G-RISK) that quantifies glaucomatous damage from color fundus photographs, identify individuals with glaucoma and reveal novel associations with genetic data from a large population-based dataset?

Objectives:
1. To validate G-RISK’s ability to quantify glaucomatous damage using UK Biobank’s extensive fundus photograph dataset by comparing continuous G-RISK scores to ground truth data, such as self-reported glaucoma or related clinical metrics.
2. To correlate G-RISK scores with relevant metadata, including demographic, lifestyle, and clinical factors, to potentially discover unknown associations related to glaucomatous damage.
3. To explore associations between G-RISK scores and genetic data available in the UK Biobank to investigate potential genetic contributions to glaucomatous damage.

Scientific rationale:
Glaucoma is a progressive optic neuropathy and a leading cause of irreversible blindness. Automated quantification of glaucomatous damage can help to lower the large number of undetected cases in the general population. G-RISK is an extensively validated deep learning model that quantifies glaucomatous damage from color fundus photographs.
This project aligns with UK Biobank’s mission to leverage innovative tools for advancing disease understanding and improving health outcomes.

Dissemination of results:
We plan to share the results of our research through a variety of channels to ensure broad accessibility and impact:
1. Academic publication in a peer-reviewed journals focused on ophthalmology, AI in healthcare, and genetics
2. Scientific conferences like the Association for Research in Vision and Ophthalmology (ARVO)
The pre-trained deep learning model will not be publicly shared, nor will it be finetuned on UK Biobank data, complying to the AI Policy.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/applying-deep-learning-to-understand-disease-genotype-relationships

Applying Deep Learning to Understand Disease-Genotype Relationships

Last updated:
ID:
33751
Start date:
30 May 2019
Project status:
Current
Principal investigator:
Dr Kyle Farh
Lead institution:
Illumina Inc., United States of America

A major outstanding question is how nature and nurture work together to impact human health. To bring new insights to this question, we are applying new artificial intelligence techniques to decipher the complex interactions between genes and environment. The broad data from the UK biobank is ideal for this approach, because artificial intelligence methods can find discoveries on their own in an unsupervised fashion, without requiring specific hypothesis. In line with the spirit of the UK Biobank’s purpose of expanding biomedical research and discoveries, we will be publishing our discoveries to help advance human clinical genetics.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/applying-deep-machine-learning-methods-for-understanding-the-complex-relationship-between-genes-and-phenotype

Applying (deep) machine learning methods for understanding the complex relationship between genes and phenotype

Last updated:
ID:
46146
Start date:
30 May 2019
Project status:
Current
Principal investigator:
Professor Jaegyoon Ahn
Lead institution:
Incheon National University, Korea (South)

Aims, scientific rationale and duration: The purpose of this study is to find the (deep) machine learning model to predict phenotype such as height, hair color and eye color, which are mostly from multi-factorial inheritance. Many studies have shown the influence of genetic variations to these phenotype. We think applying various deep machine learning models to large number of genomic samples can predict these phenotype more accurately, and would like to try various (deep) machine learning models for three years.

Public health impact: Our proposal will provide people a means to know or understand the effect of their genetic factors to their appearance, and eventually, they can be provided with more proper non-genetic information. For example, people can understand nutrients or exercises for increasing the height or loosing weight, by comparing the predicted height or weight by genetic factors, non-genetic factors and all the factors. Our proposal also will help people to live more healthy life. For example, people who have the genetic factors which increase the risk of obesity can be notified their health condition in advance.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/applying-integrative-multi-omics-and-clinical-analysis-to-define-patient-stratifying-biomarkers-in-diabetic-kidney-disease

Applying integrative multi-omics and clinical analysis to define patient-stratifying biomarkers in diabetic kidney disease

Last updated:
ID:
98468
Start date:
22 December 2022
Project status:
Current
Principal investigator:
Dr Andrew Parton
Lead institution:
MultiOmic Health Limited, Great Britain

Diabetic Kidney Disease (“DKD”), is a frequent complication of type 2 diabetes mellitus. It is characterized as a diabetes-related decline in kidney function that often results in end stage renal disease, dialysis and/or kidney transplantation. In addition, this disease often reduces the ability of the physicians to treat other diabetes-related conditions such as cardiovascular complications, owing to a decrease in the kidney’s capacity to remove harmful substances and medications from the bloodstream and into the urine.
So far, there have been few attempts to examine the early to middle stages of DKD in patients, as most of the time this condition is identified very late in the development of the disease. Hence, there has been little understanding of what triggers DKD and what drives the progression of the disease. We also know that not all DKD patients progress at the same trajectory but there are no current ways to predict or group these patients.
Recent advances in methods to analyse molecular-level disease factors combined with the advancement so-called artificial intelligence-enabled computational techniques can now enable scientists to examine this disease in much more detail and understand what drives it – an opportunity to reduce the impact of DKD on patients and their families.
This research project will investigate the biological and molecular factors that influence the early and middle stages of DKD, using data available from the UK Biobank. Assuming we are successful, the results of this project will help the broader scientific and medical community to improve the health of DKD patients and to drastically upgrade their quality of life.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/applying-machine-learning-to-metabolic-diseases-biomarker-verification-pathway-analysis-predictive-model-enhancement-and-personalized-medicine-with-explainable-ai

Applying machine learning to metabolic diseases: biomarker verification, pathway analysis, predictive model enhancement, and personalized medicine with Explainable AI

Last updated:
ID:
324631
Start date:
17 December 2024
Project status:
Current
Principal investigator:
Professor Rui Wang-Sattler
Lead institution:
Helmholtz Zentrum Munchen, Germany

Using data from the UK Biobank (UKBB), we aim to improve our ability to predict metabolic diseases utilizing advanced deep-learning models. These models will help generate new incident cases, balance the data, and improve the accuracy of our predictions. With the extensive omics data available from the UKBB, we plan to identify and validate candidate biomarkers associated with metabolic diseases, providing insights into the underlying mechanisms and causes of these conditions.

Our project will also focus on developing risk prediction models using Explainable AI (XAI). This approach will not only predict the risk of metabolic diseases but also provide personalized interpretations, helping both clinicians and patients understand the factors contributing to the risk.

The project is planned to last for three years, with the possibility of an extension.

This research will have several public health benefits. It will train young researchers in handling larger datasets and using sophisticated analytical techniques. The findings are expected to lead to high-impact publications and deepen our understanding of metabolic pathways and disease mechanisms. Ultimately, the project aims to contribute to clinical practice by improving disease prediction and prevention, thereby enhancing public health outcomes.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/applying-polygenic-risk-scores-to-the-spectrum-of-diverticular-disease

Applying Polygenic Risk Scores to the Spectrum of Diverticular Disease

Last updated:
ID:
100702
Start date:
30 June 2023
Project status:
Current
Principal investigator:
Mr Thomas E E Ueland
Lead institution:
Vanderbilt University, United States of America

Diverticular disease is a widespread source of chronic health burden. We know that a healthy lifestyle is important to preventing progression of diverticular disease, and we are beginning to understand more about some of the genetic risk factors involved. One way of translating genetic information to tools that are useful for doctors and patients is through a polygenic risk score, which combines many small genetic effects linked with a disease into a single susceptibility score. Specifically in diverticular disease, there has been some evidence that a polygenic risk score may be able to help identify individuals with more of a tendency to get the disease. Before this can be used, we must understand whether the polygenic risk score helps when we consider other known risk factors such as lifestyle variables. Our aim is to study how a polygenic risk score performs in risk prediction for diverticular disease when looking at the effects of a person’s lifestyle. This will help us in identifying a potential future role for polygenic risk scores alongside existing tools used by doctors for risk prediction.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/applying-strategies-for-lifecourse-epidemiology-by-combining-genetics-the-principles-of-mendelian-randomisation-and-longitudinal-observational-methods

Applying strategies for lifecourse epidemiology by combining genetics, the principles of Mendelian randomisation and longitudinal observational methods

Last updated:
ID:
76538
Start date:
17 January 2022
Project status:
Closed
Principal investigator:
Ms Grace Marion Power
Lead institution:
University of Bristol, Great Britain

Often diseases diagnosed in adulthood have physiological antecedents that begin in early life. Gaining a better understanding of how the timing of exposures at different stages in the lifecourse influence health outcomes is key to identifying when the effects of risk factors driving health inequalities may be reversed through lifestyle or environmental modifications. A lifecourse approach crucially investigates the contribution of early and later life exposures together, to identify risk and protective mechanisms across the lifespan. Separating the effects of risk factors at different stages of the lifecourse is challenging, particularly due to the influence of confounding factors; variables that influences both the exposure and outcome of interest, causing a spurious association. Mendelian randomisation (MR) exploits the random assortment of genetic variants, independent of other traits, to enable analyses that largely mitigate against distortions resulting from confounding and reverse causality, which afflict epidemiological observational research. However, whilst it has been shown that genetic associations may arise from the direct effects of the same inherited genetic variants at different stages of the lifecourse, there are only a handful of studies that have incorporate a lifecourse approach using genetics and applying the principles of MR.

This study sets out to assess disease risk in later life by employing instruments used to separate exposures at different stages in the lifecourse to elucidate modifiable pathways at critical time points. The more we understand about causal risk factors across the lifecourse, the more effective our interventions will be.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/appraising-causal-relationship-of-air-pollution-exposure-in-pregnancy-and-low-birth-weight

Appraising Causal Relationship of Air Pollution Exposure in Pregnancy and Low Birth Weight

Last updated:
ID:
73238
Start date:
14 December 2021
Project status:
Current
Principal investigator:
Professor Xia Yankai
Lead institution:
Nanjing Medical University, China

Aims: We aim to assess the causal relationship between gestational exposure of air pollution and low birth weight. Scientific rationale: Air pollution adversely affects health outcomes. Particularly, women are vulnerable to environmental exposure in pregnancy, which is a critical window for maternal and neonatal health. Evidence is accumulating that maternal PM2.5 exposure is related to low birth weight. From the perspective of mechanism, fine particulate matter constituents might transfer across the placental barrier and consequently lead to intrauterine growth restriction. Additionally, the consistent association has been observed in our independent cohort study. We will use data obtained from the UK Biobank to identify causal association between gestational exposure of air pollution and low birth weight via Mendelian randomization (MR). Project duration: The duration of this project will be three years, from 2021 to 2024. Public health impact: Our study could find causal detrimental effects of air pollution on low birth weight, providing critical information and knowledge that can be used by regulatory agencies, decision makers, and others to put programs and policies in place to limit our exposures to air pollution, thereby preventing or reducing the likelihood that a disease or other negative health outcome would occur and improve well-being of women and children.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/are-genetic-variants-associated-with-social-connectedness-in-uk-biobank-participants-also-associated-with-individual-variation-in-motor-function

Are genetic variants associated with social connectedness in UK Biobank participants also associated with individual variation in motor function?

Last updated:
ID:
95886
Start date:
30 May 2023
Project status:
Current
Principal investigator:
Dr Jennifer Cook
Lead institution:
University of Birmingham, Great Britain

The aim of this research is to investigate which genes contribute to differences in social behaviour and movement in people. We will also explore whether there is an overlap between genes linked to social behaviours and movement in humans and those previously identified in honeybees. We will use data collected from a large group of adults who took part in the UK Biobank project alongside honeybee data from the University of Illinois will identify key genes that have been used and reused to build social behaviours throughout the course of evolution.
Previous work has shown that certain aspects of sociability in humans and honeybees are underpinned by similar gene sets and that humans social support is a protective factor against a range of mental and physical health issues. Thus, understanding more about the genetic basis of social behaviour may help create new biologically-targeted strategies to support individuals who struggle most to access social support. We believe furthering our knowledge about the genetic underpinning of social behaviour could also help in identifying those who are most at risk of poor health outcomes relating to difficulties with utilising social support. This information could also be used to develop biologically-informed treatments to boost people’s capacity to be socially connected. This would then have a tangible public health benefit to society by ensuring more individuals who suffer with ill-health are able to tap into a readily available resource for greater health and wellbeing – that of connecting with other people.
Additionally, the information we will gain from our research will allow us to better understand how difficulties with both social and motor functioning might occur in people with neurological and psychiatric disorders. This in turn may provide a valuable opportunity to develop new therapies which can be used to treat patients with conditions such as Parkinson’s diseases. Additionally, this knowledge has the potential to highlight bio-markers that could be regularly measured in these patients to assess changes in social and motor functions. This in turn could aid in developing targeted therapeutic interventions to help regulate the biological systems governed by the affected genes.
This study is being undertaken by a research group at the University of Birmingham who have a strong commitment to public dissemination of research findings and intend to share the results from this work through peer-reviewed publications in scientific journals, presentations at scientific meetings and interactive workshops at public engagement events.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/are-individuals-with-inflammatory-arthritis-at-higher-risk-of-falling-and-do-they-have-poorer-bone-health-as-assessed-by-heel-ultrasound-and-dual-energy-x-ray-absorptiometry-scanning

Are individuals with inflammatory arthritis at higher risk of falling, and do they have poorer bone health, as assessed by heel ultrasound and dual energy x-ray absorptiometry scanning?

Last updated:
ID:
11983
Start date:
3 August 2015
Project status:
Closed
Principal investigator:
Professor Elaine Dennison
Lead institution:
University of Southampton, Great Britain

Individuals who have inflammation of their joints are thought to be at greater risk of breaking bones for several reasons; they may have thinner bones either because of their arthritis, or medication that has been prescribed to help control it, and/ or they may be more likely to fall over if the arthritis involves the joints in their lower legs. This study will look at how commonly falls are reported, and consider the measurements of heel ultrasound that have been made, in people who did or did not report a history of arthritis. These results will inform us regarding the risks faced by arthritis patients to their bone health. The findings will inform strategies to educate patients and their physicians, and will lead to prevention of fracture associated comorbidity. Because fall reduction and medication to improve bone density are currently managed through different patient pathways, it will be important to consider the excess attributable risk of both falls and low bone density in such a large, well characterised population. Although only heel ultrasound measurements are available currently, analyses would include DXA based analysis when these results are available in a subset of Biobank. We request data on self-report of arthritis, including medication use; blood results (ESR; CRP; RF) that may help in further defining groups of patients with some types of arthritis (gout, some types of rheumatoid arthritis),falls history and heel ultrasound measurements (and DXA measurements when available). We will consider risk of falls, and poorer bone health in those with or without arthritis, before and after adjusting for other factors that might be important (for example age, sex, other medical problems, cigarette and alcohol use, physical activity, medication use). Details of statistical analyses can be provided if helpful. We would like to make a phased application with cross-sectional and longitudinal analyses. We request access to the full cohort; in addition we would like to request access to the subset in whom DXA measurements have been performed when available, in addition to linkage to HES.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/are-there-any-advantages-of-hip-resurfacing-and-unicompartmental-knee-arthroplasty-on-physical-activity-levels-in-hip-and-knee-arthritis-patients-a-uk-biobank-analysis

Are there any advantages of Hip Resurfacing and Unicompartmental Knee Arthroplasty on Physical Activity Levels in Hip and Knee Arthritis Patients? A UK Biobank Analysis

Last updated:
ID:
194406
Start date:
18 June 2024
Project status:
Current
Principal investigator:
Dr Omar Musbahi
Lead institution:
Imperial College London, Great Britain

Arthritis is a condition that causes swelling, pain, and stiffness in the joints, which are the places where two bones meet, like your knees, hips, or fingers. Osteoarthritis is “wear and tear” type, where the protective cushioning called cartilage at the ends of your bones wears down over time. It primarily affects the joints that hold the most weight of the body, which is the hip and knee. Treating arthritis in the hips and knees focuses on managing pain, improving joint function, and slowing down joint damage by medication and physical therapy. When those treatments have not helped, surgery is an option. In the hip, this includes totally replacing the hip with artificial parts to mimic the hip’s natural motion (total hip arthroplasty), or by giving your hip joint a new surface rather than replacing it entirely (hip resurfacing). Similar principles are followed with total knee arthroplasty and partial/unicompartmental knee arthroplasty. These surgeries vary in type and technique. Our study is designed to show whether surgery is the most effective option for improving the activity levels of patients with arthritis, and if so, which specific type of surgery has the most benefits. We will be assessing how active patients are after having one of the four different surgery types, comparing them to individuals with arthritis who have not had any surgery, as well as healthy individuals. This comparison will use UKBiobank’s data, collected through a medical device patients wear post-surgery. This device tracks how many steps they take, how fast they are moving, and their overall activity levels. Our aim to understand which specific hip or knee surgery helps patients become more active.

Staying active is crucial for health, but arthritis can significantly affect a person’s mobility and quality of life. Our study will compare activity levels post-different surgeries to identify the most effective one, helping the decision-making of patients and surgeons about maintaining an active life after surgery. This project, spanning 24 months, has significant implications for public health. By understanding how different hip and knee surgeries in patients with arthritis impact activity levels, we will be able to understand which surgery best help arthritis patients resume normal activities and also saves money in the healthcare system. The results from the study could also provide updated information on the guidelines that doctor follow and offer more a personalised and effective care to patients.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/arguing-with-physical-activity-data

Arguing with Physical Activity Data

Last updated:
ID:
349719
Start date:
10 June 2025
Project status:
Current
Principal investigator:
Mr Adam Gould
Lead institution:
Imperial College London, Great Britain

Aims:
1. To explore how physical activity data of patients with neurological conditions can support clinical decision-making by the use of interpretable machine learning.
2. To assess how physical activity measures compete with or complement other patient assessment methods, namely with imaging and patient reported outcome questionnaires.
Scientific Rationale:
As both wearable technology and artificial intelligence permeates society, the intersection of both offers great potential in patient health assessments. In particular, patients with neurological diseases must often undergo assessments that are time-consuming, physically exhausting and expensive. Furthermore, patients in developing countries often do not have access to necessary equipment. Physical activity data from wearables, analysed and explained by machine learning techniques, can provide an objective, accurate, longitudinal and relatively inexpensive solution.
Current research showcases the potential for physical activity data as an assessment tool of patients with neurological diseases. Often, as disease progresses, patients show reduced physical functioning. However, this alone is insufficient for deciding how best to utilise this data and current research fails to fill this gap. Most importantly, we must focus on how best to communicate the analysis of physical activity to clinicians and patients.
Our research aims to empower clinicians to make better decisions based on predicted patient outcomes using physical activity data with a combination of other measures, including imaging and patient reported outcomes where possible. Predictions are explained using argumentative reasoning which has been shown to be an effective technique for communicating comprehensive decision outcomes, particularly in situations with conflicting information or when new information requires updating existing conclusions, as will often be the case with long-term monitoring of patients.
Project Duration:
Three Years.
Public Health Impact:
We aim to build on evidence of physical activity in healthcare assessments. We plan on creating concrete examples with techniques publicly available and published in peer-reviewed journals and at respected conferences. We will showcase how this data can be best used and communicated to both clinicians and patients.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/artificial-intelligence-ai-and-radiomics-for-cardiac-and-brain-magnetic-resonance-based-imaging-analysis-artifact-reduction-quality-assessment-and-interaction-between-cardiac-and-brain

Artificial intelligence (AI) and radiomics for cardiac and brain magnetic resonance based imaging analysis: artifact reduction, quality assessment and interaction between cardiac and brain

Last updated:
ID:
134960
Start date:
6 December 2023
Project status:
Current
Principal investigator:
Mr Yinan Wang
Lead institution:
Shanghai Jiao Tong University, China

Cardiac and Brain magnetic resonance is a high-quality and non-invasive imaging tool to study heart and brain structure along with function.

Benefit from fast-developing computer technology, artificial intelligence (AI) has become more and more common in daily life. Deep learning (DL) as an important part of AI, showed excellent performance in many tasks. In the field of medical image analysis, DL already exhibited perfect results in some areas, however, its disadvantages were also obvious. One of the most import drawback is that DL is hard to explain. Meanwhile, another method, called ‘radiomics’, provide an more explicit way to analyse and explain medical images.

Therefore, in this study, I intend to use the combination of AI, radiomics to analyse CMR images. I will focus on the following questions: 1) detect low quality images and try to improve them; 2) design several automatic and accurate segmentation models to facilitate image analysis; 3) comparing models’ performance and human-level performance; 4) extract and select radiomics features from images; 5) combining clinical information, radiomic features and DL models to study the interaction between brain and heart under different diseases; 5) finally, incorporate all parts together.

According to my observation and previous experiment on a small dataset. We hypothesized that combination of AI and radiomics could provides a more accurate and robust analysis framework .

This study will be conducted for 2.5-3 years with an excellent server. I hope that our proposed system could facilitate medical images quality control/improvement and clinical decision.

The codes for image processing, model architecture will be open-sourced with my paper and thesis. Also, I hope the manual segmenation of CMR images (me, 2 years of CMR experience) could be used in other research projects, and I will upload this part of data to UK Biobank if necessary.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/artificial-intelligence-ai-applications-to-medical-images-to-measure-and-stratify-the-effects-of-obesity-and-metabolic-imbalances-on-the-human-body-function

Artificial intelligence (AI) applications to medical images to measure and stratify the effects of obesity and metabolic imbalances on the human body function.

Last updated:
ID:
474792
Start date:
24 March 2025
Project status:
Current
Principal investigator:
Dr Benito de Celis Alonso
Lead institution:
Benemérita Autonomous University of Puebla, Mexico

“We intend to develop software to complement the diagnosis and quantification of fat deposits in the body and metabolic imbalances (iron concentration). Then, through the analysis of MR brain scans, assess the effects of these values on neuronal connectivity (DTI) and functional connectivity (Resting States networks (RS)). As data is available for the same subjects over time, assess if the software developed was able to predict through biomarkers values of obesity and iron concentration on volunteers with time.
Workflow is as follows:
1. With IA, quantify the concentrations of iron in liver, spleen and pancreas. All this based on MR images of these organs.
2. Automatically quantify abdominal, subcutaneous or infiltration in organ of fat (liver, pancreas and spleen). Also consider extremities and any other body parts of relevance. We would have to develop different AI Machine Learning ML applications to do this automatically. Work would have to also focus on segmentations of tissues and all based on MRI images. If possible, correlate with other anthropometric data and blood samples. Correlation could be complemented with the use of neuronal networks to find relationships between these variables.
3. Analyse RS, DTI, and other MR modalities to associate results with the parameters obtained in points 1 and 2. Assess how they influence brain function. An in-detail study of connectivity and correlation of these graphic parameters with iron and fat would also be looked for, trying to find which brain areas are mostly affected and how. As before correlations or neuronal networks would be used to find relationships between these variables.
4. Find in points 1, 2 and 3, biomarkers that can predict the evolution of accumulation of fat, iron and changes in brain function and structure with time. This using AI and ML techniques. This would serve as a diagnostic and prediction tool. This would be possible due to the information over time found for a same patient in Biobank.”


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/artificial-intelligence-based-approaches-to-identify-biological-behavioural-and-environmental-determinants-of-healthy-longevity

Artificial intelligence-based approaches to identify biological, behavioural, and environmental determinants of healthy longevity

Last updated:
ID:
208480
Start date:
17 May 2025
Project status:
Current
Principal investigator:
Professor Daniela Capello
Lead institution:
University of Eastern Piedmont Amedeo Avogadro, Italy

The objective of the present study is to discern the biological, social and environmental factors and their possible interplay in the ageing process. This objective holds relevance given the recent and sustained growth in the global elderly population. A substantial proportion of them experiences frailty and deal with multiple chronic conditions, exerting a profound influence on their quality of life as well as on the responsibilities of their caregivers. Understanding the complex interactions of these determinants of ageing is pivotal in gaining a comprehensive insight into the mechanisms underlying the achievement of healthy longevity. In this context, AI demonstrated efficacy in detecting pertinent patterns in intricate, nonlinear data, allowing the building of causal models, extracting the most important features, and identifying biological targets and mechanisms of the ageing process. The outcomes of this research will help to design interventions to promote healthy longevity and early identification of individuals at higher risk of accelerated ageing and future health issues. Due to the complex nature of the required analyses to meet our project’s objectives, it is anticipated that the project will span 36 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/artificial-intelligence-driven-autism-spectrum-diagnosis-and-subtype-classification

Artificial Intelligence-Driven Autism Spectrum Diagnosis and Subtype Classification

Last updated:
ID:
498212
Start date:
27 June 2025
Project status:
Current
Principal investigator:
Professor Kwok Wing Tsui
Lead institution:
Chinese University of Hong Kong, China

1.Research Questions
How can AI enhance ASD diagnosis accuracy?
What are the most effective AI models for ASD subtype classification?
How do genetic and behavioral contribute to AI-driven ASD diagnosis?
2.Objectives
Develop AI tools for early and precise ASD diagnosis.
Classify ASD subtypes using AI to improve personalized treatment strategies.
Integrate diverse data sources to enhance AI model performance.
3.Scientific Rationale
Unmet need for early ASD diagnosis and intervention
Potential of AI in pattern recognition within complex medical data
Synergy of interdisciplinary data (genetics, behavior, imaging) in AI models
4.To disseminate our model and findings, we aim to publish a research paper and make our model publicly available on GitHub, thereby contributing to the enhancement of diagnostic accuracy in the field.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/artificial-intelligence-enabled-risk-stratification-in-glaucoma

Artificial Intelligence-enabled Risk Stratification in Glaucoma

Last updated:
ID:
406148
Start date:
5 September 2025
Project status:
Current
Principal investigator:
Mr Yichuan Liang
Lead institution:
University of Sydney, Australia

Glaucoma is the leading cause of irreversible blindness worldwide. It is a term that describes an array of eye conditions that share a characteristic pattern of damage to the nerves transmitting visual information from the eye. The speed at which glaucoma worsens is highly variable. Current treatments can slow or arrest the worsening of glaucoma, but any existing damage will most likely persist. It is therefore important to predict the risk of glaucoma development and worsening before and after diagnosis such that tailored treatment may be planned to maximally preserve vision, however, we lack clinical tools that accurately predicts the risk of developing glaucoma or glaucoma worsening for the individual patient.
This project aims to utilise the power of Artificial Intelligence (AI) to develop a tool that can accurately predict the risk of developing glaucoma or glaucoma worsening. AI models have great potential in this area, as they can receive a wide range of inputs such as images of the eye and other informations from blood tests or medical records and perform complex computations on these inputs to generate a risk prediction that can be interpreted by the clinician. Additionally, information about the general health of the patient as well as the genetic makeup of the patient have also been linked to glaucoma, so they will be incorporated into the AI models and used for predicting risks of glaucoma worsening.
This project is expected to be conducted over three years, and its outcomes will be communicated via publications and presentations at scientific meetings. We expect this study to contribute new information about what might cause glaucoma and make glaucoma worse. We also expect this study to provide a technical foundation for an AI-enabled clinical risk assessment tool which could be used in eye clinics and further improve the outcome of individuals who might develop glaucoma or high-risk glaucoma patients especially from the early stages of disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/artificial-intelligence-for-automatic-measurement-of-heart-function-in-heart-cmr

Artificial Intelligence for Automatic Measurement of Heart Function in Heart CMR

Last updated:
ID:
78983
Start date:
21 June 2022
Project status:
Current
Principal investigator:
Dr Yanran Wang
Lead institution:
University of Science and Technology of China, China

Artificial Intelligence (the simulation of human intelligence processes by machines) has a clear advantage over humans for heart disease diagnosis. Artificial Intelligence has been successfully applied in echocardiographic images, but not cardiac magnetic resonance imaging (CMR), which is the golden standards for many heart diseases. In this project, we aim to apply Artificial Intelligence on heart CMR to enable automatic and objective heart function measurements. These heart function measurements are fundamental for diagnosis, and guidance of treatment in patients with heart diseases. Our hypothesis is that fully automated AI-enabled measurements on heart CMR can make use of the whole information from heart CMR, provide more objective assessment, and improve diagnostic accuracy.

The project will be conducted for three years and intend to use as many heart CMR scans as available to satisfy the need for automatic heart disease diagnosis. We hope that the proposed automated Artificial Intelligence on heart CMR can free doctors from tedious redundant work, improve healthcare quality, and make healthcare more accessible.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/artificial-intelligence-for-deep-phenotyping-and-target-discovery-in-cardiovascular-disease-including-heart-failure

Artificial intelligence for deep phenotyping and target discovery in cardiovascular disease including heart failure

Last updated:
ID:
116292
Start date:
25 January 2024
Project status:
Current
Principal investigator:
Dr Ramneek Gupta
Lead institution:
Novo Nordisk Research Centre Oxford Ltd, Great Britain

Cardiovascular disease (CVD) is a broad group of diseases affecting the heart and blood vessels. Heart failure (HF) is a disease where the heart is unable to carry out its normal function of pumping blood around the body. HF is a long-term condition that often gets worse, and although the symptoms can be managed, there is currently no cure. There are many different causes of CVD, and the severity of the disease and its progression varies across patients. There can be differences in how CVD patients are treated, and how patients respond to treatment. Currently in HF, a measure of the heart’s ability to pump blood, called left ventricular ejection fraction (LVEF), is used to assign HF patients into groups. However, we know that there are still high levels of variation in patient groups based on LVEF.
We aim to better understand the variation across patients in CVD, including HF. This will allow us to improve the management and treatment of CVD patients, and to develop new drugs for specific groups of CVD patients. We will use clinical records from CVD patients, combined with genetic information, medical images of the heart and information about physical activity, to model CVD and its progression to other diseases. We will use advanced methods such as artificial intelligence and computational simulations, to understand the risk factors and mechanisms of CVD. We aim to improve our understanding of why some patients progress rapidly and develop other diseases, and why some patients may not respond to treatment. We hope to identify new sub-groups of CVD patients that are clinically and biologically similar to each other, and clinical markers of these groups. Using these improved CVD sub-groups, we aim to discover new drug targets for CVD that may treat specific causes of the disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/artificial-intelligence-for-early-screening-and-risk-stratification-of-systemic-diseases-using-multimodal-retinal-images

Artificial Intelligence for Early Screening and Risk Stratification of Systemic Diseases using Multimodal Retinal Images

Last updated:
ID:
911747
Start date:
28 September 2025
Project status:
Current
Principal investigator:
Dr Carol Cheung
Lead institution:
Chinese University of Hong Kong, China

Research Questions: 1. Can deep learning (DL) models detect systemic diseases by using retinal images, demographic data, and risk factors? 2. Can DL models predict the future risk of developing systemic diseases using baseline retinal images and risk factors? 3. What specific retinal biomarkers are associated with systemic diseases?

Objectives: Aim 1. To develop DL models capable of detecting systemic diseases and predicting future disease risk by integrating retinal image-derived biomarkers with established risk factors. Aim 2. To investigate associations between retinal biomarkers extracted from retinal images and a broad spectrum of systemic diseases.

Scientific Rationale: The retina offers a unique, non-invasive window into systemic health. Sharing embryological origins and microvascular architecture with vital organs, retinal alterations often precede clinical manifestations of systemic diseases by years. Fundus photography (FP) captures vascular morphology changes linked to hypertension, diabetes, and stroke risk. Optical coherence tomography (OCT) reveals neuronal layer thinning associated with preclinical neurodegenerative diseases. These modalities provide highly accessible, automatable, and cost-effective tools for large-scale screening and risk stratification.
With the global burden of chronic diseases rising, innovative approaches for early detection and risk stratification are critical, especially since many conditions remain asymptomatic until irreversible damage occurs. Deep learning (DL) excels at identifying subtle, complex patterns beyond human capability directly from images.
This project will harness advanced AI techniques, designed to integrate multimodal data, and the advantages of UK Biobank with its unprecedented scale, longitudinal design, and rich linked health data to learn complex patterns indicative of systemic pathologies directly from retinal images.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/artificial-intelligence-for-using-multi-modal-data-to-improve-the-identification-and-prediction-of-diseases-in-the-uk-biobank

Artificial intelligence for using multi-modal data to improve the identification and prediction of diseases in the UK Biobank

Last updated:
ID:
100739
Start date:
27 June 2023
Project status:
Current
Principal investigator:
Dr Kai Huang
Lead institution:
Sun Yat-sen Memorial Hospital, Sun Yat-sen University, China

Scientific rationale: Cardiovascular diseases (CVDs) remain the leading cause of death worldwide accounting for ~32% of all global deaths, with the expectation that this number will rise to > 23.6 million deaths annually by 2030. Also, the morbidity and mortality of cancers and other chronic diseases, and severe or acute diseases are rapidly growing and becoming prominent obstacles to increasing life expectancy. These diseases are a growing concern, and new techniques are needed to promote healthcare centers worldwide to appropriately manage them. In our research project, artificial intelligence (AI) will be used to provide key technological support to address clinical problems. Based on deep learning on the comprehensive data, AI algorithms with well-performed accuracy will play an important role in the early prevention, identification, treatment and prognosis on diseases. Furthermore, with the assistance of AI, the individualized diagnosis and treatment process will be achieved, and clinical problems encountered in the medical process could be settled.

Aims: In this research, we seek to: (1) Incorporate multiple levels of omics data (genomics, transcriptomics, proteomics, metabolomics, etc.) to improve the identification and prediction of diseases. (2) Develop artificial intelligence models for early detection and prognosis prediction of diseases using Biobank imaging data. (3) Investigate the influence of genetic information and environment risk factors to the occurrence and development of diseases based on Biobank comprehensive individualized data. (4) Develop artificial intelligence models to identify disease risks using multi-modal data across different levels of biology.

Project duration: The duration of the project will be for 36 months.

Public health impact: Our AI systems are ultimately designed for clinical application and to settle unmet clinical demands comprising early prevention, diagnosis and treatment of CVDs, cancers or other chronic diseases, and severe or acute diseases. Our AI systems based on UK Biobank cohort study have the potential to achieve global generalization and then benefit the patients. Furthermore, our research will provide a favorable opportunity for drug discovery and development process, which will eventually contribute to the rapid development of effective drugs.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/artificial-intelligence-machine-learning-for-cardiovascular-diseases-in-multiscale-modeling

Artificial intelligence/machine learning for Cardiovascular Diseases in Multiscale Modeling

Last updated:
ID:
76333
Start date:
2 September 2021
Project status:
Closed
Principal investigator:
Dr Ching-Heng Lin
Lead institution:
Linkou Chang Gung Memorial Hospital, Taiwan, Province of China

According to the WHO report, 17.9 million people die each year from cardiovascular diseases (CVDs), an estimated 31% of all deaths worldwide, CVDs also has placed a heavy burden on patients and society. Although various risk factors that related to CVDs have been identified in many previous clinical studies, here still remain challenges to identify predictive features that can improve CVDs prediction and diagnosis model. Artificial intelligence/machine learning (AI/ML) techniques are known to be excellent at identifying important features and making a prediction. AI/ML with multiscale modeling approach is a rapidly growing field. It can help identify new targets and treatment strategies, and inform decision making for the benefit of human health. Multiscale modeling is developing models that represent multiple different scales (population, individual, organ, cellular, or molecular level) and how they interact with each other. Integrating AI/ML and multiscale modeling can be a powerful tool to help researchers to understand complex CVDs towards developing more accuracy predictive and diagnosis model and optimizing treatments of CVDs. This study aims to develop an integrated CVDs diagnosis and outcome prediction platform with AI/ML assisted analytical tools that can find the clinically meaningful pattern of CVDs and provide diagnosis, recommended treatment options with predicted outcome to better identify CVDs patients for better outcomes.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/artificial-intelligence-methods-applied-to-cardiovascular-mri-segmentation

Artificial intelligence methods applied to cardiovascular MRI segmentation.

Last updated:
ID:
44929
Start date:
3 July 2019
Project status:
Current
Principal investigator:
Mr Youssef Skandarani
Lead institution:
University of Burgundy, France

We aim to help physician to achieve more accurate, reliable and rapid diagnosis of the different conditions that may affect a patient, through the analysis of the magnetic resonance images with methods based on machine learning techniques.It would assist the cardiologists in the interpretation of the acquired images and, since they nowadays spend a lot of their time performing image post-processing, enable them to save time for other tasks (particularly meet the patients). The management and the follow-up of the patients will be more streamlined by considering objective biomarkers automatically obtained from validated methods.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/artificial-intelligence-research-for-cardiac-electrical-biomarker-identification

Artificial intelligence research for cardiac electrical biomarker identification

Last updated:
ID:
89372
Start date:
30 September 2022
Project status:
Current
Principal investigator:
Dr Joon-myoun Kwon
Lead institution:
Incheon Sejong Hospital, Korea (South)

Aims:
Analysis of changes in electrocardiogram will enable the early diagnosis and prevention of various diseases. Several lines of evidence support the use of ECG(Electrocardiogram) data to find association with critical diseases. However, studies that have aimed to predict diseases through changes in electrocardiogram with strong accuracy and state-of-the-art techniques is lacking. We aim to investigate the usefulness of electrocardiogram, such as rhythm and patterns to evaluate their relations with our target outcomes, and to identify a novel cardiac electrical biomarker that can be used to predict changes in patients.
Scientific rationale:
According to a recent study, AI(Artificial Intelligence)-enabled ECGs can be used to detect heart-related diseases and events.
Currently, the relationship between electrocardiogram and disease is a mostly extracted under human knowledge based on conventional medical evidence. However, there is a possibility of finding novel electrophysiological biomarkers by analyzing bigdata using artificial intelligence methods.
Artificial Intelligence analysis methods can analyze waveform data itself, such as an electrocardiogram. It has advantage to explore the relation between the electrocardiogram and the disease through automated feature extraction (a machine learning method that does not require manually curated features).
Project duration: about 3 years
– 6 months: data preparation and build a database
– 3~4 months: data pre-processing and evaluation of existing association
– 1 year: discovery of a novel cardiac electrical biomarker using the deep learning model
– 6 months: subgroup analysis
– 3~4 months: summary and supplement of research results
Expected impact on the public health:
Our study will contribute a novel cardiac electrical biomarker that can be used to predict changes in electrocardiogram. The marker can be used for the early detection and prevention of various diseases. Specifically, the cardiac electrical biomarkers can be used to monitor high-risk patients with frequent diseases non-invasively and continuously using mobile devices. Furthermore, an intervention strategy can be established to prevent or delay the progression of diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/artificial-intelligence-supported-data-harmonization-and-analysis-for-the-identification-of-prognostic-factors-related-to-balance-gait-disorders-and-risk-of-falls

Artificial intelligence supported data harmonization and analysis, for the identification of prognostic factors related to balance/gait disorders and risk of falls

Last updated:
ID:
101577
Start date:
7 September 2023
Project status:
Current
Principal investigator:
Professor Doris-Eva Bamiou
Lead institution:
University College London, Great Britain

In this project we will review the UK Biobank data to find which factors or characteristics may contribute to falls or increase the risk of falls. The reason for the project is that there appears to be a gap in the scientific knowledge regarding the factors that may increase an individual’s risk of falls – throught the interaction of and combined effect of the chronic conditions, psychological and behavioural factors on the individual. We aim to review medical history and the chronic conditions, as well as genetic test results and imaging from the UK Biobank data – to find out which such factors may predicit falls or poor balance which may affect balance and reduce level of activity in the individuals. In addition to reviewing the risk factors for worsening of well-being and the risk of falls, we will look at the data to find out any factors that may predict response to treatment or may cause treatment side effects, and how likely an individuals is to use new technologies for treatment (and continue with such new treatment). We also aim to find out which factors may help individuals with their balance, lower their falls risk and help them keep active. We will aim to then combine the results of the data analysis from this project with the results from other projects – including the results of data analysis from other data sets. We hope that by doing so we then be able to put together recommendations for use in clinical setting – for a better and more targeted (specific) treatment that can be offered to patients in the future.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/artificial-neural-network-based-integration-of-genomic-metabolomic-environmental-and-clinical-information-to-predict-the-risk-of-cerebrovascular-specific-diseases-in-patients-with-colorectal-cancer

Artificial neural network-based integration of genomic, metabolomic, environmental and clinical information to predict the risk of cerebrovascular-specific diseases in patients with colorectal cancer

Last updated:
ID:
89551
Start date:
25 August 2022
Project status:
Current
Principal investigator:
Professor Guo-Dong Chen
Lead institution:
University of South China, China

Colorectal cancer (CRC) is one of the most prevalent diseases and the second leading cause of death worldwide. With the advance in diagnosis and therapy of CRC, patient death due to this cancer reduces greatly, putting mortality from other non-cancer diseases an unignorable concern for this population at present. Among them, cerebrovascular-specific disease (CVSD) constitutes a major factor contributing to the mortality in CRC patients. However, the current evidence concerning the CVSD occurrence in CRC is limited. A recent Surveillance, Epidemiology, and End Results (SEER)-based cohort study from our group indicated an increased CVSD mortality in CRC and also found several clinical parameters linked to this outcome. However, due to the incomplete patient information in SEER database, our study only addressed the relationship between some clinicopathological factors and CVSD events in CRC, which may not provide adequately accurate information on this correlation. As a continuation of our previous work, the present project will further analyze the genomic, metabolomic and environmental predispositions to CVSD in CRC patients by using the more homogeneous, comprehensive, and complete data in the UK Biobank database. More importantly, we also seek to develop and validate an artificial neural network-based model to predict the risk of CVSD in this population. The estimated project duration is three years. We believe that our project results will provide helpful information in uncovering the potential mechanism of CVSD development in CRC and can also be useful in guiding risk stratification and preventive intervention optimization of CVSD in the patients.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assesments-of-joint-effect-of-polygenic-disease-risk-scores-and-modifiable-risk-factors-on-major-chronic-diseases-and-mortality

Assesments of Joint Effect of Polygenic Disease-risk Scores and Modifiable Risk-factors on Major Chronic diseases and Mortality

Last updated:
ID:
17712
Start date:
15 August 2016
Project status:
Current
Principal investigator:
Professor Nilanjan Chatterjee
Lead institution:
Johns Hopkins University, United States of America

The aim of the research is to use data from UK Biobank prospective cohort study to estimate joint risk of multiple common disease conditions, including cancer, type-2 diabetes, cardiovascular diseases, and overall mortality, associated with GWAS generated polygenic disease-risk scores and modifiable risk-factors such as smoking, BMI, alcohol and physical activity. The analysis will allow understanding of how the potential impact intervention on modifiable risk-factors, may or may not vary by individual’s genetic risk-profiles. The proposed research will generate valuable information regarding whether genetic information could be useful for targeting certain primary prevention efforts for risk-factor intervention that cannot be applied to the general population for cost and other burdens. For subjects in the UK Biobank cohort, polygenic risk-score (PRS) for major chronic diseases will be constructed based on published literature on susceptibility SNPs and their disease odds-ratios. Data from UK Biobank will be then used to estimate absolute risk of different disease endpoints and mortality in population strata defined by polygenic risk scores and modifiable risk-factors. For evaluating combined endpoint like overall mortality or overall cancer incidence, the disease-specific PRSs will be combined to form a composite PRS. Further, to understand the potential causal effect of intervention of modifiable risk-factors, like BMI, a Mendelian Randomization approach will be used to estimate absolute risk-reduction parameters associated with BMI for the different outcomes within strata defined by the disease associated PRS variables. These `instrumental` variable derived absolute-risk reduction parameters will then be then compared with more direct epidemiological estimate of the same parameters for the assessment of consistency of results across two types of analyses. All subjects in the full cohort with or without genotype data


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assess-the-causal-relationship-between-dietary-and-lifestyle-behaviors-and-chronic-diseases-during-the-aging-process-particularly-the-occurrence-and-development-of-metabolic-disorders

Assess the causal relationship between dietary and lifestyle behaviors and chronic diseases during the aging process, particularly the occurrence and development of metabolic disorders.

Last updated:
ID:
1035127
Start date:
28 September 2025
Project status:
Current
Principal investigator:
Dr Peihao Wu
Lead institution:
Nanjing Medical University, China

Research Question: How do dietary and lifestyle behaviors impact the development and progression of metabolic disorders during aging? What specific dietary components and lifestyle factors are most strongly associated with these chronic diseases in older adults?
Objectives: (1) To understand the causal relationship between dietary and lifestyle behaviors and the occurrence and progression of metabolic disorders in the aging population. (2) To identify key dietary components and lifestyle factors that contribute to the onset and progression of these chronic diseases. (3) To examine how changes in these behaviors over time influence disease trajectories in older adults.
Scientific Rationale: Metabolic disorders are a significant health concern among older adults, contributing to reduced quality of life and increased healthcare costs. As people age, their bodies undergo physiological changes that can increase the risk of developing chronic diseases. Dietary and lifestyle behaviors play a crucial role in modulating these risks. By understanding the specific dietary components and lifestyle factors that influence metabolic health, we can develop targeted interventions to improve the health and well-being of older adults. This study aims to fill gaps in our knowledge by providing comprehensive insights into the interplay between diet, lifestyle, and metabolic health in aging populations. The findings will be instrumental in shaping public health policies and personalized health recommendations for older adults.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-allele-phasing-algorithms

Assessing allele phasing algorithms

Last updated:
ID:
20613
Start date:
30 June 2016
Project status:
Closed
Principal investigator:
Gerton Lunter
Lead institution:
University of Oxford, Great Britain

To identify genetic mutations related to disease, a `genome wide association study` (GWAS) is often conducted. As a technical first step of any such study, it is necessary to assign each heterozygous variant to the paternal or maternal chromosome, a process called `Phasing`.

I have developed a new phasing algorithm for large cohorts, and I would like to test this algorithm on the 150,000 genotyped individuals in the UK Biobank.

This test would be very similar to that used in http://biorxiv.org/content/early/2015/12/18/028282, which used the same data set. Health-related, and in the public interest:

The proposed project aims to improve a necessary, technical step in GWAS studies. Improvements would make GWAS studies more efficient, and easier to conduct as cohort sizes grow. This would simplify the discovery of disease-related associations with genetic variants, establishing the health connection. The public interest is served by simplifying the research, potentially speeding up and/or reducing the cost of research. I would apply the new algorithm to the data, and compare the results with existing algorithms. This is explained in detail here: http://biorxiv.org/content/early/2015/12/18/028282 I am applying for the full data set of genotypes (152,725 individuals).


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-and-evaluating-polygenic-scores-of-complex-phenotypic-traits-in-admixed-populations

Assessing and evaluating polygenic scores of complex phenotypic traits in admixed populations

Last updated:
ID:
74348
Start date:
3 August 2021
Project status:
Current
Principal investigator:
Professor Michel Satya Naslavsky
Lead institution:
Albert Einstein Israelite Hospital, Brazil

In a given population, it is challenging to predict how characteristics (from physical differences to having or not a disease) occur based on each individual genetic composition (mutations carried by each person). Most prevalent conditions, such as hypertension and dementia are considered complex, because they are caused by a combination of environmental factors and genetic components, which are hard to be detected since their individual contribution (each mutation) is very small. Large initiatives such as the UK Biobank improve this detection. However, in different populations these mutations may have different effects (and different mutations also), which are still largely unknown, since most projects enroll individuals of European descent. As a result, when scientists calculate the combined effect of mutations in one group (such as the UK Biobank participants) to predict a disease, it is likely that the same list of mutations will not have the same performance in a different population, especially when they are admixed. In this 3-year project, we wish to compare how characteristics and diseases are found in UK Biobank and in a Brazilian group of individuals, which were whole genome sequenced and are genetically admixed. After that, we will test if the combinations of mutations that predict a disease in UK Biobank can also be used in the Brazilian group. Finally, we will test if combining other types of information such as knowledge on how genes are activated and how gene products interact within the context of the diseases can be used to improve the combined effect of mutations and recover part of the predictive qualities observed in UK Biobank to other populations (such as Brazilians). Comprehending specific features that define how to increase performance when transferring these mutation across populations may contribute to direct public health applications such as monitoring higher risk groups and to understanding novel genetic interactions that promote the manifestation of complex diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-and-mitigating-major-adverse-liver-outcomes-among-patients-with-metabolic-associated-steatotic-liver-diseases-risk-factor-analysis-based-on-multi-dimensional-data

Assessing and Mitigating Major Adverse Liver Outcomes among patients with metabolic-associated steatotic liver diseases: risk factor analysis based on multi-dimensional data

Last updated:
ID:
177053
Start date:
4 February 2025
Project status:
Current
Principal investigator:
Dr Yun Shen
Lead institution:
Pennington Biomedical Research Center, United States of America

Our research project aims to understand and prevent serious liver problems in people with liver diseases linked to metabolic issues, like obesity and diabetes. We plan to figure out what factors increase the risk of these liver problems, how to predict them, and the best ways to prevent them. Liver diseases related to metabolic issues, like too much fat in the liver, are becoming more common. These conditions can lead to serious health problems, including liver failure. However, it’s tough to predict who will develop these severe problems and how to prevent them. Our project will use a mix of genetics, health data, and lifestyle information to get a clearer picture of these diseases. This approach is important because it looks at the whole picture, including how genes and lifestyle choices like diet and exercise affect liver health. We will use data from the UK Biobank, a large health database, to study people with these liver diseases. We’ll look at their genetic information, health records, and lifestyle habits. We’ll also use a method called Mendelian Randomization, which helps us understand if a certain factor (like a diet) actually causes liver problems or not. Plus, we’ll use advanced computer techniques (machine learning) to analyze this data and predict who might be at risk for serious liver issues. Our research can have a big impact on public health. By understanding what causes serious liver problems in these patients, we can develop better ways to prevent and treat these conditions. This could mean fewer people suffering from severe liver diseases, less strain on healthcare systems, and more effective treatments. Also, our findings could help educate people about the risks of these liver conditions and how to avoid them, leading to healthier communities. n summary, our project aims to shed light on a growing health issue by combining genetics, health data, and computer analysis. Our goal is to improve the prevention and treatment of serious liver diseases related to metabolic issues, benefiting both individuals and the wider public health. The project is expected to take 36 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-association-between-genome-sequence-variants-biomarkers-proteins-and-immune-mediated-diseases

Assessing association between genome sequence variants, biomarkers, proteins, and immune mediated diseases

Last updated:
ID:
85823
Start date:
14 July 2022
Project status:
Current
Principal investigator:
Mr Ward Ortmann
Lead institution:
HIBio, Inc, United States of America

This research project aims to identify genetic variants and biomarkers that affect the chances that someone develops an immune-mediated disease (including ‘long COVID’). Previous genetic studies have already identified many loci and/or mutations that are implicated in disease risk or severity. However, the UK Biobank, by its sheer size and scope, offers additional opportunities for refining our understanding of disease risk, which may lead to new insights on drugs or treatments for disease. Non-genetic factors, such as age, sex and body mass index, are also known to affect disease risk. We also plan to combine genetic insights with these non-genetic risk factors to better predict an individual’s risk profile for immune-mediated disease development or progression. This project will last at least three years, and may help with the development of new drugs or drug targets.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-association-of-risks-and-prognoses-between-mental-disorders-e-g-depression-anxiety-bipolar-disorder-schizophrenia-and-cardiovascular-diseases-cvd

Assessing association of risks and prognoses between mental disorders (e.g., depression, anxiety, bipolar disorder, schizophrenia) and cardiovascular diseases (CVD).

Last updated:
ID:
608779
Start date:
30 April 2025
Project status:
Current
Principal investigator:
Dr Mengyang Jia
Lead institution:
Jinan University, China

Mental disorders (e.g., depression, anxiety, bipolar disorder, schizophrenia) and CVD are deeply interconnected global health challenges with substantial individual and societal impacts. Despite extensive research, the mechanisms underlying their bidirectional relationship remain inadequately understood. Utilizing the UK Biobank dataset, this study will examine how mental disorders-such as depression, anxiety, bipolar disorder, and schizophrenia-contribute to the onset, progression, and prognosis of CVD, focusing on pathways like inflammation, autonomic dysregulation, and neuroendocrine disturbances. Additionally, we will investigate the interplay between genetic susceptibility and modifiable factors, including psychosocial stress, sleep dysfunction, and lifestyle behaviors, in shaping the risk for both conditions. Emphasis will also be placed on identifying shared biomarkers across inflammatory, neuroendocrine, and metabolic axes to improve early detection, risk stratification, and individualized therapeutic strategies. By integrating mental health into cardiovascular disease research, this study aims to advance the emerging field of psychocardiology, fostering precision medicine approaches to reduce the global burden of these comorbid conditions and enhance patient outcomes. The results of this research would be used as doctoral thesis and would be published on scholarly journals.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-covariate-mediated-polygenetic-risk-scores-for-mortality

Assessing covariate-mediated polygenetic risk scores for mortality

Last updated:
ID:
104381
Start date:
12 July 2023
Project status:
Current
Principal investigator:
Mr Yoann Colin
Lead institution:
Australian National University, Australia

Technological advances have made genetic data easier to obtain at large scale, holding the promise of an individualised approach to genetic-informed health care. That said, the same genetics that informs treatment may also be predictive of risk, which can be notably used to predict life expectancy more accurately. Here we use large scale genetic data to model mortality risk and assess its contribution relative to more conventional mortality factors such as health and family history.

The public health impact of the proposed research is multitudinous. The most notable impact of whole DNA incorporation into models for mortality prediction will allow us to find out whether using entire DNA sequencing is too course for increasing accuracy of life expectancy predictions or whether it can in fact add to current models at play in the health sector. In this sense, DNA can potentially allow individuals to better evaluate their mortality and thereby optimise their own health outcomes through lifestyle choices based on better informed decision making processes. Additionally, the research would allow health professionals to give more tailored advice to their patients, thus allowing such individuals to take ownership of their health with greater confidence and potentially making choices that are more effective in prolonging their wellbeing.

As a preliminary study to future models and investigations, we anticipate a 12-month project duration.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-frequency-distribution-of-the-hht-mutations

Assessing frequency distribution of the HHT mutations

Last updated:
ID:
329910
Start date:
4 February 2025
Project status:
Current
Principal investigator:
Dr Alexey Lugovskoy
Lead institution:
Diagonal Therapeutics Inc., United States of America

Aim: To analyze and quantify the frequencies of various mutations associated with Hereditary Hemorrhagic Telangiectasia (HHT), aiming to provide a comprehensive understanding of the genetic landscape of this disorder.

Scientific Rationale: HHT, affecting over 1.4 million people globally, is a complex genetic disorder with significant variability in its genetic underpinnings. The ClinVar database identifies over 900 genetic variants in key genes (ENG, ACVRL1, SMAD4) linked to HHT. Despite this, there is a substantial gap in the literature regarding the prevalence of each mutation per gene. Our project aims to fill this gap by categorizing these mutations as either rare or common, thus enhancing our understanding of the disease’s genetic basis.
Project Duration: 12 months
Public Health Impact: This research will greatly impact public health. By identifying the most common mutations associated with HHT, our findings will guide future clinical studies, aiding in the development of more precise diagnostic tools and targeted therapeutic strategies. This project represents a significant stride towards better understanding the genetic landscape of HHT and pave the way for advanced clinical interventions, ultimately benefiting the large patient population affected by this disorder.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-genomic-determinants-underlying-disease-pathogenesis-and-clinical-course-by-joint-analyses-of-the-uk-biobank-and-genomics-england-datasets-in-the-national-genomic-research-library-ngrl

Assessing genomic determinants underlying disease pathogenesis and clinical course by joint analyses of the UK Biobank and Genomics England datasets in the National Genomic Research Library (NGRL)

Last updated:
ID:
196939
Start date:
15 October 2024
Project status:
Current
Principal investigator:
Dr Loukas Moutsianas
Lead institution:
Genomics England, Great Britain

Genomics England partners with the NHS to enable whole genome sequencing diagnostics. Our aim is to improve genomic healthcare and contribute to scientific research.

We focus on identifying new genetic factors related to disease, particularly rare diseases and cancer. We do that by utilising statistical methods for analyzing large-scale genomic data and other modalities such as protein abundance and gene expression data from proteomics and transcriptomics assays, respectively. By doing that, we aim to better understand how diseases develop and progress. In the process, we refine our clinical interpretation pipelines for the identification of disease-causing variants on individuals from diverse genetic backgrounds and improve healthcare for everyone.

We will use our National Genome Research library alongside the UK Biobank multimodal data. Our aim is to support our joint missions to advance modern medicine and treatment through scientific discovery.

We aim to identify novel determinants of human health and biomarkers of clinical course, compare statistical association results across disease and trait domains for replication and undertake joint analysis, and share novel discoveries with the scientific community.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-improvement-of-chronic-diseases-through-hypothetical-intervention

Assessing Improvement of Chronic Diseases through Hypothetical Intervention

Last updated:
ID:
104967
Start date:
5 February 2024
Project status:
Current
Principal investigator:
Professor Jingkai Wei
Lead institution:
University of Texas (UT Health), United States of America

Chronic conditions, such as cardiovascular disease and dementia, have become a huge burden of public health. A series of risk factors, such as physical inactivity, unhealthy diet, smoking, depression, hearing and vision impairment, high blood pressure, glucose, and cholesterol have been identified to be related chronic diseases. Although observational studies have found that some healthy actions against these risk factors, such as healthy diet and physical activity are related to a lower risk of these chronic diseases. However, evidence from these studies cannot be considered causal, and randomized trial is the type of study that may draw causal inference, but randomized trials may suffer from the issue of feasibility. For example, a randomized trial typically lasts for less than 5 years, but some intervention may last longer to show effectiveness. Under this situation, causal analysis of observational studies become the optimal choice. In this proposed study, we will study whether we can reduce the risk of chronic diseases in population with lifestyle change (diet, physical activity, smoking cessation, sleep improvement), vascular medication (antihypertensive medication, statins) and improvement of vascular risk factors (blood pressure, cholesterol, glucose), improving sensory impairments, and improving mental health. If the study is successfully conducted with significant findings, it can guide clinicians and public health professionals with reasonable actions on reducing burden of chronic disease in population.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-improvement-of-non-communicable-diseases-based-on-lifestyle-intervention-environmental-factors-and-genetic-factors

Assessing improvement of non-communicable diseases based on lifestyle intervention, environmental factors and genetic factors

Last updated:
ID:
283055
Start date:
5 November 2024
Project status:
Current
Principal investigator:
Mr Yan Li
Lead institution:
Guangxi Zhuang Autonomous Region People's Hospital, China

Non-communicable diseases such as cardiovascular disease, cancer, chronic respiratory disease, and diabetes have become a huge burden on public health. Various risk factors, including lack of physical activity, an unhealthy diet, smoking, depression, hearing and vision impairments, and environmental pollution, have been associated with non-communicable diseases. Adjustable lifestyle factors, including diet, physical activity, alcohol consumption, smoking habits, and sleep patterns, play a crucial role in lowering the incidence of these diseases. Although previous studies reported that some healthy lifestyles against these non-communicable diseases. Nevertheless, the evidence derived from these studies cannot establish causality. While randomized trials are capable of drawing causal inferences, they may encounter feasibility challenges. Consequently, elucidating key risk factors and developing prediction models based on these factors holds significant potential for identifying high-risk populations, thereby enhancing their overall healthy life expectancy and minimizing unnecessary medical expenses. In this project, we aim to investigate the potential for mitigating the risk of non-communicable diseases in the population through lifestyle modifications (such as dietary adjustments, increased physical activity, smoking cessation, and alcohol withdrawal), and enhancements in vascular risk factors (such as management blood pressure, cholesterol levels, and glucose) and environmental factors (such as heavy metal pollution), and genetics (SNPs, etc.). Furthermore, we will evaluate those factors to enhance mental well-being. Given that non-communicable diseases arise from a combination of genetic, behavioral, and environmental risk factors, Consequently, as part of this project, our strategic approach will be rooted in data-driven methodologies. Assessing and improvement of that incorporate multiple such factors theoretically offers improved predictive performance. The objective of this study is to employ appropriate statistical methods in order to uncover potential associations between the factors listed above and the occurrence of non-communicable diseases. By rigorously analyzing data, we aim to elucidate meaningful insights that can inform preventive strategies and enhance our understanding of disease etiology by leveraging multidimensional data from the UK Biobank.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-liver-health-through-morphometry

Assessing liver health through morphometry

Last updated:
ID:
58840
Start date:
27 April 2021
Project status:
Current
Principal investigator:
Dr Jonathan Karl Riek
Lead institution:
BioTel Research, LLC, United States of America

The aim of this study is to determine if the amount of fibrosis in the liver can be determined by looking at the shape and size of the liver, the spleen and the hepatic (liver) blood vessels.
The liver can be divided into three sections – the right lobe, the left medial lobe and the left lateral lobe. The two left lobes can be further divided into a top and bottom (or superior and inferior) section. The right lobe can be divided into 4 sections by dividing it into a top and bottom section, and a front and back (anterior and posterior). Many researchers have looked at how these lobes and sections change in size and shape when there is fibrosis present in the liver. Generally, the left lateral lobe gets larger and the right lobe gets smaller as the amount of fibrosis increases. The spleen also tends to enlarge with increasing fibrosis in the liver. Additionally, the surface of the liver gets rougher, the hepatic veins tend to get narrower, and a notch sometimes appears in the bottom back section of the right lobe of the liver. Most of this information can be extracted from the MR images obtained in this study. Combining this morphometry information with the measured fat and the corrected-T1 should provide a relatively complete picture of the overall liver health without having to take a biopsy.
A multi-stage deep learning approach will be used to identify the spleen, the liver, and the different segments within the liver. For the liver and spleen identification, the first stage will identify which images contain the organ of interest. From those images, the second stage will identify the boundary of the organ based on learnings from manual identification of the organ.
For the individual segments in the liver, the first stage will identify images that contain anatomical landmarks that can be used to divide the liver into the different segments. A second stage will take these identified images and attempt to draw boundary lines between the segments. The final stage will divide the liver into the different segments based upon the boundary lines that were identified.
Once these regions are identified, the volumes and relative sizes can be calculated as described in the literature to assess fibrosis/cirrhosis risk.
It is anticipated that this research project will take approximately 24 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-metformin-use-in-t2d-patients-to-suppress-autoimmune-diseases

Assessing metformin use in T2D patients to suppress autoimmune diseases

Last updated:
ID:
60130
Start date:
4 May 2020
Project status:
Closed
Principal investigator:
Dr Daniel H Robertson
Lead institution:
Indiana Biosciences Research Institute, United States of America

Summary: Type 2 Diabetes is projected to become the biggest epidemic disease in the world. Studies have shown correlation between T2D and different autoimmune diseases. People diagnosed with prediabetes and diabetes are prescribed with metformin. As metformin has immune modulatory effects, therefore identifying the cohorts with T2D+autoimmune and metformin will help to understand whether metformin has evidence to delay autoimmune diseases (and which) in the broader population as capture in this real-world dataset.

Aims: (i) Evaluate demographics, clinical, medications, and socio-determinants of T2D+autoimmune disease type cohorts that are treated with and without metformin.
(ii) Identify key features that can be used as markers for each cohort using ANOVA/MANOVAs.
(iii) The methodology developed in (i) and (ii) using the UK Biobank will be implemented on another similar INPC USA dataset.
(iv) The key finding using UK Biobank and INPC USA dataset will be compared/contrast.
(v) The key features will be tested as markers using ML.

Scientific Rationale: As metformin is prescribed as first line of drug for the treatment of prediabetes and diabetes and studies have reported immune-modulatory effects of metformin for T2D patients. In this project we plan to study the cohorts that are treated with/without/ before/after metformin as preventative measures for autoimmune disease. Completion of the study can show if metformin can really help to control the progression of T2D to autoimmune disease. On comparing two different population cohorts across world can help to understand the prevalence and the similarity/differences. The key features identified using statistical approaches will be evaluated as markers using different machine learning methods.

Project Duration: We estimate a total 2 years to complete the project.

Public Health Impact: As it is projected by 2045 nearly 10%-12% of the world population will be affected with diabetes. Moreover, studies have shown the association of autoimmune diseases with T2D. Both these disease types effect the quality of life and incur lot of medication expenditure. This study can assist better understanding if metformin has evidence to delay onset of specific autoimmune diseases (and which) and in which patient populations. Based on these results it could lead to (1) identification of new markers of risk for specific autoimmune disease, (2) evidence for further study of metformin in specific autoimmune disease and patient populations, or (3) additional electronic (Machine Learning) tools to identify patients at risk of developing autoimmune disease assisting in clinical monitoring and treatment.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-multimorbidity-and-polypharmacy-patterns-in-chronic-respiratory-diseases-and-their-impact-on-clinical-outcomes

Assessing Multimorbidity and Polypharmacy Patterns in Chronic Respiratory Diseases and Their Impact on Clinical Outcomes

Last updated:
ID:
611404
Start date:
11 March 2025
Project status:
Current
Principal investigator:
Mr Changcheng Shi
Lead institution:
Chinese Academy of Medical Sciences &Peking Union Medical College, China

Research question
By 2030, the burden of chronic respiratory diseases (CRDs) like chronic obstructive pulmonary disease (COPD) and asthma is projected to increase due to aging and environment problem. Multimorbidity and polypharmacy in CRD patients are common, yet the knowledge about their impact on clinical outcomes and underlying patterns are still limited.
Objective
This study aims to leverage the comprehensive UK Biobank dataset to investigate multimorbidity and polypharmacy patterns in CRD patients. Specifically, the objectives are to:
1. Identify common multimorbidity patterns in CRD patients and assess their prevalence and distribution.
2. Characterize polypharmacy practices and evaluate their appropriateness in the context of multimorbidity.
3. Investigate the risk factors associated with the development of multimorbidity in CRD patients, including demographic, genetic, lifestyle, and clinical variables.
4. Explore the impact of these patterns on clinical outcomes, including hospitalization rates, mortality, and quality of life.
Scientific rationale
Multimorbidity in CRD patients often leads to complex treatment regimens, increasing the risk of adverse drug events and poorer outcomes. Investigating these patterns offers an opportunity to identify actionable risk factors, improve disease management, and optimize medication strategies. By combining rich epidemiological data with advanced analytical techniques, this study will provide insights into:
1. The clustering of comorbidities and their interactions with CRD progression.
2. Patterns of medication use, highlighting potential risks of polypharmacy.
3. The risk factors contributing to the development of multimorbidity in CRD patients.
4. Causal relationships between identified patterns and clinical outcomes.
This research will offer evidence-based guidance to healthcare providers, improve treatment strategies, and ultimately enhance the quality of life for individuals with CRDs.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-novel-risk-factors-using-multimodal-data-to-enable-the-prediction-of-developing-parkinsons-disease

Assessing novel risk factors using multimodal data to enable the prediction of developing Parkinson’s disease

Last updated:
ID:
171229
Start date:
15 May 2024
Project status:
Current
Principal investigator:
Dr Thomas Payne
Lead institution:
University of Sheffield, Great Britain

Parkinson’s disease is a brain disease that results in people developing difficulties with movement such as difficulties walking, making fine movements with their fingers and also developing problems such as stiffness and tremor. Some people with Parkinson’s can also develop problems with their memory or hallucinations. Parkinson’s develops when brain cells in a very small and specific part of the brain involved in movement unfortunately degenerate and die, this area is called the ‘substantia nigra’. We currently don’t have any treatments that slow down the progression of Parkinson’s which will continue to deteriorate over years.

One problem with developing new treatments for Parkinson’s is that by the time people have developed the movement related symptoms of Parkinson’s they will frequently have actually had symptoms for years. In fact, the processes that ultimately lead to the development of Parkinson’s may begin decades before the development of symptoms. When people do develop the movement symptoms of Parkinson’s they have already lost a significant proportion (roughly 50%) of the brain cells in the substantia nigra. Therefore, to enable effective treatment of Parkinson’s we need to be able to identify people in the earliest stages of the disease to then target with any potential treatments that may slow down the progression of the disease.

We have identified a naturally occurring bile acid (ursodeoxycholic acid) as a promising treatment for slowing down the progression of Parkinson’s. We now want to investigate if other medical conditions or environmental factors that affect the bile acids in the body may affect the risk of developing Parkinson’s. After identifying any relevant new risk factors that affect the bile acid composition within an individual, we will then use a machine learning method (sometimes referred to or used interchangeably with artificial intelligence) to develop a predictive model that can look to identify individuals at high risk of being in the early stages of Parkinson’s before the development of the movement symptoms.

We would envisage this project taking between 12 and 36 months to complete. Identifying new risk factors implicated in the development of Parkinson’s may help guide the investigation and development of new treatments for Parkinson’s. A predictive model of Parkinson’s could also be utilised in identifying people who may best be targeted in future clinical trials of new drugs.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-ntrk1-snps-in-pain-disorders

Assessing NTRK1 SNPs in pain disorders

Last updated:
ID:
51933
Start date:
30 August 2019
Project status:
Current
Principal investigator:
Dr Juan Carlos Arevalo
Lead institution:
University of Salamanca, Spain

Chronic pain is becoming a huge problem in society nowadays. The constant feeling of pain has negative effects on work and personal life due to anxiety, fear, depression, sleepiness and impaired social interaction, which represent a worldwide health and social burden. Indeed, these adverse effects constitute a huge economic cost, which is estimated to be around $600 billion in the US and !300 billion in the EU annually. Thus, effective treatments are urgently needed to treat chronic pain, but in order to develop them it is necessary to understand the mechanisms underlying chronic pain. Nerve growth factor (NGF) and its receptor (NTRK1) have been directly associated as pain mediators. Numerous variants of NTRK1 have been described in the human population but their biological relevance on pain is unknown. In this project, we aim to identify if any of these NTRK1 variants is associated with any pathology related with chronic pain. To this purpose, we will take advantage of the data gathered at the UK Biobank in terms of human DNA sequences and their corresponding disease/pathology information. Our research will try to identify new variants of the NTRK1 gene related with pain syndromes. The successful outcome of the proposed work will allow to compile very relevant information that will be used to study the mechanisms by which NTRK1 affects pain and eventually the design of drugs to prevent/alleviate chronic pain.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-optimal-treatment-protocols-for-pelvic-fractures-in-motor-vehicle-accident-patients

Assessing Optimal Treatment Protocols for Pelvic Fractures in Motor Vehicle Accident Patients

Last updated:
ID:
716731
Start date:
27 March 2025
Project status:
Current
Principal investigator:
Professor Li-ping Huang
Lead institution:
Chinese PLA General Hospital, China

Research Questions:

Does early internal fixation improve survival rates within the first 30 days post-injury in patients with motor vehicaccident(MVA)-induced pelvic fractures?
How do early internal fixation, delayed fixation, and non-surgical treatments compare in terms of complication raes, pain management, functional recovery, and length of hospital stay?
What factors (e.g. fracture type, patient demographics, coexisting injuries) influence the selection and efficacy otreatment modalities for MVA-related pelvic fractures?
Objectives:

To evaluate the impact of early internal fixation on survival rates in patients with VA-induced pelvic fractures.
To compare the efficacy of early internal fixation, delayed fixation, and non-surgical treatments in terms of clinicaoutcomes.
To identify factors that influence treatment selection and patient outcomes.
To establish evidence-based guidelines for the management of MVA-related pelvic fractures.
Scientific Rationale!
Pelvic fractures resulting from MVAs are associated with high morbidity and mortality due to severe structural and vascular damage. The lack of standardized treatment protocols leads to variability in management and patient outcomes. Early internal fixation may stabilize fractures, prevent life-threatening complications (e.g., hemorrhage, shock), and improve survival rates. However, evidence comparing early fixation with delayed or non-surgical approaches is limited. This study aims to fill this gap by evaluating the efficacy of different treatment protocols,focusing on survival, complications, pain management, and functional recovery. The findings will provide evidence-based insights to optimize treatment strategies, reduce recovery time, and enhance patient outcomes, ultimatelv advancing trauma care and patient-centered recovery.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-prevalence-and-progression-of-type-2-diabetes-comorbidities

Assessing prevalence and progression of type 2 diabetes comorbidities

Last updated:
ID:
61168
Start date:
24 June 2020
Project status:
Current
Principal investigator:
Dr Daniel H Robertson
Lead institution:
Indiana Biosciences Research Institute, United States of America

Summary: Type 2 Diabetes (T2D) is projected to become the biggest epidemic disease in the world. People with T2D often progress to complex co-morbidity like cardiovascular disease, kidney disease and liver disease. These associated diseases are driving increased healthcare costs. T2D itself is driving by a complex set of biological, socio-economic, and behavior factors. Effective prevention or management of T2D improves long-term outcomes and reduced healthcare costs.

Aims: This study aims to analyze progression of T2D patients to different co-morbidities is universal or is dependent on the population i.e. social determinants, environment, diet, drugs, genotype and compare these across two geographically diverse populations.

The aim(s) of this study is to:
(1) Profile T2D patients from the UK Biobank with respect to the following co-morbidities: cardiovascular diseases, chronic kidney diseases, and liver diseases
(2) Identify potential markers that track with these comorbidity disease progression
(3) Compare/contrast these results from UK Biobank with patient population from the State of Indiana
(4) Identify specific interesting subsets of the broader patient population for further study.

Scientific Rationale: As progression of T2D patients to any of the complex co-morbidities results in the decline of patient’s quality of life and increases mortality. A similar research project is underway using data from the State of Indiana. Being able to compare and contrast the results across these diverse geographic patient populations, should be able to better identify key factors associated with socio-economic/environment from biological/behavioral.

Project Duration: It is expected this project will be completed in less than two years, depending on availability of appropriate data.

Public Health Impact: This study by cross-comparing with a non-overlapping dataset, should allow key factors to be better identified that can then be used for policies, new interventions, and/or therapeutic targets to slow the current trends and/or reduce the impact of the impacts of diabetes. Identification of these transferable or invariant factors and subsequent validation, should allow a reduction of currently predicted epidemic rates and associated healthcare costs.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-psychological-trauma-and-physical-health-outcomes-focusing-on-exposure-patterns-disease-heterogeneity-the-interaction-between-gene-trauma-environment-and-resilience

Assessing psychological trauma and physical health outcomes: focusing on exposure patterns, disease heterogeneity, the interaction between gene-trauma-environment, and resilience

Last updated:
ID:
70227
Start date:
18 October 2021
Project status:
Current
Principal investigator:
Dr Sun Jae Jung
Lead institution:
Yonsei University, Korea (South)

Our research question aims to find out whether trauma and posttraumatic stress disorder (PTSD) provoke cardiovascular disease and cognitive decline. Especially, we want to test if people with PTSD gene and brain structure react to which type of trauma and social condition. Across different types of trauma, we want to find similar trauma groups and predict the later health effect. To distinguish trauma type, 16 items related to past trauma experience (Field ID 20487-20491, 20521-20531) will be grouped pre-defined with the concept from previous literature.
In the U.K., about 3% of the adult population is known to have PTSD. We hypothesized that people with more severe trauma and PTSD symptoms would have a more harmful effect on cardiovascular and cognitive health. It is already known that people with PTSD frequently report cardiovascular diseases, and this was reported from observational data, which is hard to tell the cause and effect, absolutely. To fully understand the relationship, we need to understand more about the biological and behavioral actions that occur. Similarly, stroke, myocardial infarction, thromboembolism, and dementia were frequently reported among people with PTSD symptoms. It is already known that certain cardiovascular diseases and cognitive problems, including Alzheimer’s dementia, share common pathways in disease development.
There are many kinds of trauma, the initial exposure for PTSD, including violence from family members, natural disasters, car accidents, and war. However, it is unclear whether which type of trauma brings the worst health results after experiencing PTSD. Therefore, this study aimed to find specific trauma experience patterns and assess the different associations with later cardiovascular/ cognitive function. This study will be continued in 3 years for the analysis, interpretation, and writing of papers.
When we complete this study, it will contribute significantly to understand the physical aftermath of PTSD, including cardiovascular diseases and cognitive decline. It will also help determine which combination of given factors will bring a more severe form of cardiovascular/cognitive diseases. It will help to find the best preventive intervention among PTSD patients. Additionally, this study will make it possible to predict the risk of cardiovascular diseases and cognitive decline, which will also make it possible to distinguish people who need the help most urgently.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-relationships-between-schizophrenia-polygenic-risk-scores-and-other-medical-diagnoses-among-individuals-without-schizophrenia

Assessing relationships between schizophrenia polygenic risk scores and other medical diagnoses among individuals without schizophrenia

Last updated:
ID:
54928
Start date:
31 January 2020
Project status:
Closed
Principal investigator:
Dr Sarah Bergen
Lead institution:
Karolinska Institutet, Sweden

Our project seeks to understand the relationships between genetic risk for schizophrenia and comorbid disorders including both psychiatric disorders and other medical conditions.
Schizophrenia is a disabling mental disorder with profound personal and societal costs, substantial comorbidity resulting in markedly reduced life expectancy, and incompletely known risk factors. Most of the risk for schizophrenia comes from genetic sources, and >170 genetic risk variants have been identified. People with this disorder experience higher rates of many other psychiatric and other medical conditions which leads to much lower life expectancy for people with this diagnosis.

The relationships between genetic risk for schizophrenia and comorbid disorders remain largely unknown. This study will use information from previous genetic studies to construct genetic risk scores for schizophrenia which quantify the risk from genetic sources for each individual, whether or not they develop the disorder. We plan to use these scores to understand to what extent genetic risk for schizophrenia confers increased risk for other medical diagnoses.

The analysis on UKBiobank data will start as soon as the data is accessible. We estimate the duration including manuscript preparation to be approximately 9 months. Successful completion of this project will elucidate the clinical impact of genetic risk loci for schizophrenia beyond this disorder and offer insights into the causes of comorbidity responsible for most early mortality in schizophrenia. We anticipate this work will contribute to advancements in treatment and prevention efforts.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-risk-factors-and-developing-an-early-prediction-model-for-cancer-therapy-related-cardiotoxicity

Assessing Risk Factors and Developing an Early Prediction Model for Cancer Therapy-Related Cardiotoxicity

Last updated:
ID:
387995
Start date:
9 January 2025
Project status:
Current
Principal investigator:
Professor Zhong Zuo
Lead institution:
The First Affiliated Hospital of Chongqing Medical University, China

With advancements in cancer detection and treatment, the number of cancer survivors has significantly increased. However, cancer therapies often come with severe side effects, particularly cardiovascular toxicity. Cancer therapy-related cardiovascular dysfunction occurs in approximately 10% of patients and has a mortality rate exceeding 50%. Many cancer patients are more likely to die from cardiovascular issues than from cancer itself. Therefore, it is crucial to focus on early prediction and risk assessment of cardiotoxicity to improve prevention and management, ultimately enhancing long-term health outcomes for cancer patients.
Our project aims to identify risk factors and develop an early prediction model for cancer therapy-related cardiotoxicity. We also seek to uncover any genetic risk factors associated with this condition, helping clinicians stratify patients by risk level, enabling earlier interventions, and closer monitoring for those at high risk.
This three-year project will utilize data from the UK Biobank. Initially, we will integrate comprehensive data on cancer patients, including environmental and behavioral factors, laboratory results, imaging data, omics, and genetic information. By analyzing these diverse data sources, we aim to discover the risk factors that contribute to the onset and progression of cardiovascular disease following cancer therapy. Once identified, we will select the most relevant markers to develop assessment models tailored for various clinical scenarios. These models will be created using both traditional statistical methods (such as Cox regression and logistic regression) and machine learning techniques. Additionally, our research will delve into the mechanisms behind cancer therapy-related cardiotoxicity, potentially identifying new therapeutic targets.
This research underscores the need for heightened attention to cardiovascular risks in cancer patients. By providing a method for early prediction and risk assessment, our work will significantly impact the prevention and management of cancer therapy-related cardiotoxicity. Early identification of high-risk individuals will allow for timely interventions, potentially reducing morbidity and mortality rates among cancer patients. Our findings will also contribute to a better understanding of the underlying mechanisms of cardiotoxicity, paving the way for improved treatments and patient care strategies.
In summary, this project will offer valuable insights into the risk factors of cancer therapy-related cardiotoxicity and provide practical tools for early prediction, ultimately enhancing the quality of life and survival rates for cancer patients.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-risk-for-type-2-diabetes-using-uk-biobank

Assessing risk for type 2 diabetes using UK Biobank

Last updated:
ID:
27291
Start date:
31 May 2017
Project status:
Closed
Principal investigator:
Dr Kimberley Smith
Lead institution:
University of Surrey, Great Britain

We want to determine how various diabetes risk and protective factors might differ in their impact on diabetes incidence based on the baseline level of diabetes risk as determined with the Leicester Diabetes Risk Score. In the first cross sectional phase of this study we will examine how psychological, behavioural and social factors differ based on baseline diabetes risk level. The second phase will use longitudinal data to determine whether psychological, behavioural, functioning and social factors increase/decrease the risk of incident diabetes when we stratify for baseline diabetes risk score. This research will allow us to conduct research that will help us to better identify people at high risk of developing diabetes, and what factors may be most protective in reducing the risk of diabetes. It is in the publics interest to have research that helps them to better understand diabetes risk and how we can best alleviate this risk. Phase 1: We will remove any person with diabetes at baseline. We will then split the sample up according to diabetes risk score (low, increased, moderate, high). We will then look at how behavioural (e.g., activity), psychological (e.g., depression), functioning (e.g., grip strength) and social (e.g., social networks) factors are associated with each level of diabetes risk.
Phase 2: We will look at the incidence of diabetes across each level of baseline diabetes risk. We will then examine the proportional increase/decrease in risk that results from each behavioural, psychological, functioning and social factor. Full cohort


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-risks-and-prognoses-for-ocular-and-chronic-systemic-illnesses

Assessing risks and prognoses for ocular and chronic systemic illnesses

Last updated:
ID:
564893
Start date:
21 October 2025
Project status:
Current
Principal investigator:
Dr Yuxiang Hu
Lead institution:
Nanchang University, China

The research project is meticulously crafted to explore the complex realm of ocular diseases, recognizing their profound impact on global health, which includes contributing to visual impairment and blindness, and imposing a substantial societal burden. Existing epidemiological and clinical research has established connections between ocular diseases and systemic factors, while genetic studies have highlighted a genetic contribution. However, the interplay with environmental factors remains a significant area of uncertainty. The extensive dataset from the UK Biobank offers an unparalleled opportunity to analyze these intricate relationships. Through the application of various statistical methodologies and the adjustment for confounding variables, our research aims to enhance our understanding of the etiology of ocular diseases, potentially leading to innovative preventive and therapeutic approaches that can mitigate the burden of these conditions. Initially, our focus is on investigating the impact of systemic conditions such as diabetes, hypertension, autoimmune disorders, and cardiovascular diseases on the onset, progression, and severity of a range of ocular conditions, including glaucoma, cataracts, macular degeneration, diabetic retinopathy, hypertensive retinopathy, and retinopathy of prematurity. Subsequently, we intend to examine the complex interplay between genetic predispositions and environmental factors, such as smoking, diet, and physical activity, in the causation of ocular diseases. Finally, we seek to identify robust and clinically applicable biomarkers that can facilitate early detection, precise diagnosis, and personalized interventions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-association-between-inflammatory-marker-c-reactive-protein-genetic-score-predisposition-nsaids-use-and-pancreatic-cancer-risk-and-survival

Assessing the association between inflammatory marker c-reactive protein, genetic score predisposition, NSAIDS use and pancreatic cancer risk and survival

Last updated:
ID:
71177
Start date:
19 October 2021
Project status:
Current
Principal investigator:
Dr Jeanine M Genkinger
Lead institution:
Columbia University, United States of America

Pancreatic cancer is the seventh leading cause of cancer death in both men and women, but more importantly, ranks high as one of the deadliest cancers. Inflammation has been hypothesized to be one of the main pathways to cancer development and is consistently associated with many cancers. While a number of studies have found an increased risk of pancreatic cancer with inflammation, other studies found no association. These conflicting studies vary in many aspects to our proposed study – only pancreatic cancer was examined; only one study out of five included anti-inflammatory drugs use, and no studies utilized a numeric score that represents the genetic predisposition to inflammation (genetic risk score). Additionally, no studies to date have examined the association between an inflammation risk score and risk for pancreatic cancer. This study aims to address these gaps and examine this novel construct. We will assess the association between inflammation, risk score and medication use with risk of pancreatic cancer. We will calculate ratios and include relevant variables such as baseline information, education, height, smoking, alcohol, etc. This study will take approximately 16 months and will result in findings that aim to clarify and contribute to previously relevant literature on inflammation, anti-inflammatory drug use and pancreatic cancer risk. These results may be clinically relevant for pancreatic cancer prevention and help elucidate how inflammation, genetics and use of anti-inflammatory drugs contribute to one of the deadliest cancers.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-association-between-water-intake-and-all-cause-mortality-as-well-as-hydration-related-health-outcomes-in-uk-biobank-participants

Assessing the association between water intake and all-cause mortality as well as hydration related health outcomes in UK Biobank participants.

Last updated:
ID:
73012
Start date:
7 October 2021
Project status:
Current
Principal investigator:
Miss Amy Rodger
Lead institution:
University of Glasgow, Great Britain

Water is essential for survival, however, the impact of low water intake on health is under-researched. The human body constantly losses a large amount of water (e.g., through urination, sweating) and can only produce and store a limited amount of water. Due to the body’s high water loss and low production/storage of water, water must be replaced by water intake throughout the day. However, in the UK a large proportion of men and women are have low water intake. Therefore, research on the impact of low water intake is crucial as this could affect many people in the UK.
Recent research has found evidence that water intake is linked to numerous different health outcomes. Water intake can increase urine flow and decrease urine concentration reducing the recurrence of kidney stones and urinary tract infections. Additionally, water intake can reduce the level of hormones that have been linked to the development of Type 2 diabetes, chronic kidney disease, obesity and cardiovascular disease. Finally, there is evidence that increased water intake could reduce the risk of bladder cancer. However, as this area of research is relatively new, more studies are needed to assess the link between water intake and these health outcomes. This project aims to assess whether increased water intake reduces the instance of health outcomes such as urinary tract infections, Type 2 diabetes, kidney disease, obesity, cardiovascular disease and bladder cancer.
The health outcomes we will study affect many people in the UK and cost the UK economy a significant amount of money every year. For example, 5.26% of people in the UK had Type 2 diabetes in 2014 and diabetes accounts for approximately 10% of NHS expenses. If increasing water intake could potentially lower the risk of these health outcomes it is important to research this using the UK Biobank and inform the public accordingly. Informing the public is crucial, especially since current public health messages are largely silent on the implications of underhydration and low water intake.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-associations-between-mental-health-and-cardiometabolic-diseases

Assessing the Associations between Mental Health and Cardiometabolic Diseases

Last updated:
ID:
938760
Start date:
20 August 2025
Project status:
Current
Principal investigator:
Ms Lu - Wang
Lead institution:
Nanchang University, China

Research question
The prevalence of cardiometabolic diseases (CMDs), which defned as hypertension, diabetes, coronary heart disease (CHD) and stroke, is increasing rapidly. Considerable evidence has suggested that having any one of these conditions alone could increase the risk of mortality. Most studies have only focused on the cardiovascular diseases (CVD), but research on CMD is still relatively limited.
Objective
To address this gap, the present undertaking aims to employ an integrated approach, leveraging the wealth of genetics, metabolomics, proteomics, and comprehensive epidemiological data. Through multidimensional analyses, we aim to discover new risk factors, identify potential biomarkers, and understand causal relationships for various psychological disorders and cardiometabolic diseases.
Scientific rationale
Psychiatric disorders are associated with cardiometabolic diseases, but the exact relationship is unknown. To accomplish this, we will combine multi-omics (i.e., proteomics and metabolomics) and epidemiological data to discover novel risk factors, biomarkers and provide definitive evidence for known associations of psychological disorders and cardiometabolic diseases risk reported by traditional observational studies. Our endeavor will encompass the analysis of multi-omics and epidemiological information, enabling us to unravel new risk factors, identify potential biomarkers, and comprehend the intricate web of causal relationships underpinning various psychological disorders and cardiometabolic diseases. This comprehensive exploration will include conditions such as hypertension, diabetes, CHD, stroke, depression, anxiety disorder, and more.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-associations-between-psychological-disorders-and-cardiometabolic-diseases

Assessing the Associations between Psychological Disorders and Cardiometabolic Diseases

Last updated:
ID:
812052
Start date:
28 September 2025
Project status:
Current
Principal investigator:
Professor Wei Gao
Lead institution:
Nanchang University, China

Research question
The prevalence of cardiometabolic diseases (CMDs), which defned as hypertension, diabetes, coronary heart disease (CHD) and stroke, is increasing rapidly. Considerable evidence has suggested that having any one of these conditions alone could increase the risk of mortality. Most studies have only focused on the cardiovascular diseases (CVD), but research on CMD is still relatively limited.
Objective
To address this gap, the present undertaking aims to employ an integrated approach, leveraging the wealth of genetics, metabolomics, proteomics, and comprehensive epidemiological data. Through multidimensional analyses, we aim to discover new risk factors, identify potential biomarkers, and understand causal relationships for various psychological disorders and cardiometabolic diseases.
Scientific rationale
Psychiatric disorders are associated with cardiometabolic diseases, but the exact relationship is unknown. To accomplish this, we will combine multi-omics (i.e., proteomics and metabolomics) and epidemiological data to discover novel risk factors, biomarkers and provide definitive evidence for known associations of psychological disorders and cardiometabolic diseases risk reported by traditional observational studies. Our endeavor will encompass the analysis of multi-omics and epidemiological information, enabling us to unravel new risk factors, identify potential biomarkers, and comprehend the intricate web of causal relationships underpinning various psychological disorders and cardiometabolic diseases. This comprehensive exploration will include conditions such as hypertension, diabetes, CHD, stroke, depression, anxiety disorder, and more.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-associations-between-the-use-of-common-non-cancer-medications-and-the-risk-of-cancer-at-selected-sites

Assessing the associations between the use of common non-cancer medications and the risk of cancer at selected sites

Last updated:
ID:
59281
Start date:
30 April 2020
Project status:
Closed
Principal investigator:
Dr Agnès Fournier
Lead institution:
INSERM, France

Some medications may have unexpected effects on the risk of cancers. For example, menopausal hormone therapy, which was widely used in women to alleviate menopausal symptoms such as hot flushes, has been shown to increase breast cancer risk. On the other hand, drugs such as aspirin or the antidiabetic metformin are considered as potential chemopreventive agents towards some cancer sites. However, to date, no firm conclusion has been reached for many common non-cancer medications such as antihypertensive drugs, lipid-lowering drugs, antidepressants, proton-pump inhibitors, or anti-inflammatory drugs regarding their effect on cancer incidence. Our aim is therefore to evaluate the association between these drugs and the risk of cancer incidence at selected, frequent cancer sites (breast, prostate, colon-rectum, lung, skin). Our results will provide information allowing a better appraisal of the benefit-risk ratio of common medications and may help identifying already existing drugs that could be repurposed for cancer prevention.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-causal-genetic-influence-of-polycystic-ovarian-syndrome-on-preeclampsia-risk-a-mendelian-randomization-approach

Assessing the Causal Genetic Influence of Polycystic Ovarian Syndrome on Preeclampsia Risk: A Mendelian Randomization Approach

Last updated:
ID:
125644
Start date:
5 December 2023
Project status:
Current
Principal investigator:
Dr Loc Thai Huu Tran
Lead institution:
Hope Research Center, Viet Nam

Title: “Is There a Genetic Link Between Polycystic Ovarian Syndrome and Preeclampsia? A Study Using UK Biobank Data”

Aim: The goal of this study is to find out if there is a genetic link between Polycystic Ovarian Syndrome (PCOS) and preeclampsia, a condition that causes high blood pressure in pregnant women.

Why is it important? PCOS is a common condition that affects women’s hormones, making it difficult for them to get pregnant. Women with PCOS are also thought to be at higher risk of developing preeclampsia, a dangerous condition that can cause serious, even fatal complications for both mother and baby. However, we don’t yet understand why these two conditions might be linked. By investigating this, we hope to provide important insights that could help prevent or better manage preeclampsia in women with PCOS.

How will we do it? We will use a method called Mendelian Randomization, which allows us to use genetic data to figure out if there’s a cause-and-effect relationship between two things – in this case, PCOS and preeclampsia. We will use data from the UK Biobank, a large study that has collected health and genetic information from around 500,000 people.

Project Duration: The project is expected to last for 2 years. In the first year, we will collect and prepare the data, and in the second year, we will conduct our analysis and interpret the results.

Public Health Impact: If we can show a genetic link between PCOS and preeclampsia, it could help doctors to identify women who are at a higher risk of developing preeclampsia earlier in their pregnancy. It could also lead to new treatments for preeclampsia, which could ultimately improve the health and safety of mothers and babies. This project can also contribute to our broader understanding of women’s health and reproductive disorders.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-causal-relationship-between-non-communicable-disease-risk-factors-and-coronary-artery-disease-a-bidirectional-mendelian-randomization-study-across-diverse-ancestry-populations

Assessing the Causal Relationship Between Non-Communicable Disease Risk Factors and Coronary Artery Disease: A Bidirectional Mendelian Randomization Study Across Diverse Ancestry Populations

Last updated:
ID:
228325
Start date:
18 June 2024
Project status:
Current
Principal investigator:
Miss Sarah Silva
Lead institution:
London School of Hygiene and Tropical Medicine, Great Britain

Our research aims to investigate the relationship between non-communicable diseases (NCDs) and coronary artery disease (CAD) in the context of diverse ancestry populations. We are interested in understanding the causal relationship between different risk factors and CAD and understanding how, if any, the relationship differs across populations. Previous studies have shown a link between NCDs and CAD, however the causal relationship between the two, especially in diverse ancestry populations, is not yet well understood. By looking at ancestry-specific genetic factors associated with each trait through a methodology called Mendelian Randomization, we hope to uncover population-specific differences in the biological development of CAD.
The proposed data sources for our study are consortia with previously collected genome-wide association data, with our project expected to last several months, as we need time to collect, prepare and analyse the data. Understanding the relationship between different NCDs and CAD can have a significant impact on public health. If we can confirm a causal link between the two and identify ancestry-specific genetic differences in their relationship, it will help healthcare providers develop better preventive strategies and targeted interventions on a global scale. This means we can better support individuals diagnosed with different NCDs to reduce their risk of developing heart disease, ultimately improving overall health outcomes for everyone.
Our research aims not only to deepen scientific understanding but also contribute towards research focused on translating such findings into tangible benefits for individuals and communities on a global scale. Through a comprehensive exploration of the relationship between NCDs and CAD, it is with hope that our research contributes towards the development of more personalised and effective healthcare interventions, ultimately enhancing the health and well-being of individuals affected by NCDs.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-clinical-value-of-brain-age-in-different-brain-diseases

Assessing the clinical value of brain age in different brain diseases

Last updated:
ID:
110502
Start date:
1 November 2023
Project status:
Current
Principal investigator:
Dr Chang-hyun Park
Lead institution:
Ewha Womans University, Korea (South)

In contrast to chronological age, biological age is the rate at which an individual is aging physically. Brain age, as a type of biological age, is estimated for brain ageing often by using brain MRI, and it is especially crucial because cognitive decline that significantly impacts the quality of life in elderly people is mainly related to brain ageing. A model for estimating brain age can be constructed by applying different algorithms to model healthy individuals’ age as a function of various brain MRI properties. Given such a brain age estimation model, estimated brain age for an individual represents how fast or slowly your brain is aging compared with other individuals with the same age. In the context, individuals with brain diseases often show higher brain age than their chronological age, representing accelerated brain ageing for them.
Although brain age is considered a promising marker of brain ageing in healthy and pathological ageing, there would be a need to better understand the meaning of brain age in individual brain diseases or across different brain diseases. In the current project, we seek to assess the clinical value of brain age by examining differences in brain aging status represented by brain age between different brain diseases. Specifically, we are planning to (1) develop a model for accurate brain age estimation and (2) apply the brain age estimation model to different brain diseases, including Alzheimer’s disease, Parkinson’s disease, epilepsy, and stroke. Based on a better understanding of brain age according to brain diseases, we hope that brain age can be developed as a summary indicator of brain health for individuals with brain diseases as well as healthy individuals.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-covariate-specific-causal-effects-of-helicobacter-pylori-on-gastric-and-extra-gastric-diseases

Assessing the covariate-specific causal effects of helicobacter pylori on gastric and extra-gastric diseases

Last updated:
ID:
531801
Start date:
19 March 2025
Project status:
Current
Principal investigator:
Dr Wei Wang
Lead institution:
Third Military Medical University (TMMU), China

Helicobacter pylori (H. pylori) is a significant gastric pathogen, infecting over 50% of the global population. Numerous studies have linked H. pylori infection to both gastric and extra-gastric diseases. Given a considerable subject-between heterogeneity in the effects, it is crucial to assess the individual health effects of H. pylori, i.e., the effects in subjects with specific covariates. Distinguished from the previous studies that typically focused on a single covariate using a subgroup analysis and merely established the association without assessing the causal effect, this project aims to comprehensively assess the multi-covariate-specific causal effects of H. pylori infection on gastric and extra-gastric diseases by developing a novel approach which combines the flexible semi-parametric model (or nonparametric model) with the causal inference tools, such as mendelian randomization. The covariates will include sex, age, polygenic risk scores, health condition, natural environmental factors, smoking, drinking and other common lifestyle habits. This project could help identify which H. pylori-infected individuals are at a higher risk for specific diseases, ultimately improving the management and treatment of those infected.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-directionality-of-associations-between-lifestyle-factors-atopic-dermatitis-and-cardiometabolic-disease-in-the-uk-biobank

Assessing the directionality of associations between lifestyle factors, atopic dermatitis, and cardiometabolic disease in the UK Biobank

Last updated:
ID:
276825
Start date:
27 November 2024
Project status:
Current
Principal investigator:
Dr Katrina Abuabara
Lead institution:
University of California, San Francisco, United States of America

Atopic dermatitis (AD), also known as eczema, is a chronic, inflammatory skin disease resulting in a significant decrease in quality of life for patients. Patients with AD are often found to be at higher risk for developing cardiometabolic issues alongside AD, such as hypertension, which can make management of AD symptoms even more difficult. Although we know that adults with more severe AD have an increased risk of cardiometabolic outcomes, we do not currently understand the underlying reasons for this association.

Our project aims to examine how lifestyle choices, including factors like diet and smoking, affect the risk of developing AD and its related cardiometabolic issues. We will examine how the severity of AD and the amount of salt in our diet each contribute to the risk of developing cardiometabolic diseases, whether certain lifestyle choices can lead to AD or vice versa, and whether AD can lead to cardiometabolic diseases or vice versa.

To accomplish this, we will use data sourced from the health records of individuals over time, examining who develops AD and cardiometabolic diseases, and determining the statistical impact of lifestyle and salt intake on disease. We will also use a statistical model known as Mendelian Randomization to understand the causality behind the relationships mentioned above.

Through understanding these relationships better, we can provide more personalized recommendations to individuals with AD on how to prevent the development of cardiometabolic issues. This could include dietary changes, among other potential lifestyle choices, that could serve as low risk, yet highly effective prevention strategies for further development of disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-effect-of-free-water-on-brain-microstructure-in-aging-using-diffusion-mri

Assessing the effect of free water on brain microstructure in aging using diffusion MRI

Last updated:
ID:
40922
Start date:
10 October 2018
Project status:
Current
Principal investigator:
Professor Jean Chen
Lead institution:
Baycrest Academy for Research and Education, Canada

Modern society is currently dealing with an aging population, and an associated decline in brain function is posing a threat towards clinical care in a way that is not yet fully understood. The aim of this research project is to develop a better understanding of how the brain changes through healthy adult aging and to characterize these changes right when they begin, on a microstructural level, before progression into larger-scale changes. Diffusion MRI can characterize brain microstructure by measuring the microscopic diffusion of water molecules through brain tissue. However, the brain is known to shrink with age, allowing for more extracellular free water molecules to diffuse in the brains of older adults. Free water is not reflective of authentic tissue microstructure, so increases in free water at older ages can lead to biases in assessing age-effects with diffusion MRI. This research project will apply ““free water elimination” to diffusion MRI data, allowing for aging microstructure to be assessed strictly within brain tissue itself. Over 36 months of analysis, we will work to identify brain regions where microscopic diffusivity parameters become altered in healthy aging. This analysis will be conducted in both the white matter and gray matter. The gray matter is situated directly next to cerebrospinal fluid and fast-flowing blood, which are forms of free water that make the gray matter particularly susceptible to bias, and thus aging gray matter microstructure has scarcely been studied altogether with diffusion MRI. We will eliminate this free water to allow for the first ever large-scale in-vivo study of gray matter microstructural changes through healthy aging. This will furthermore allow for better distinction of pathological aging, and ultimately allow for health care systems to cater closer to the needs of its aging population.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-health-effects-of-alcohol-consumption-mechanisms-of-action-and-associated-disease-burden

Assessing the health effects of alcohol consumption, mechanisms of action, and associated disease burden

Last updated:
ID:
459270
Start date:
10 January 2025
Project status:
Current
Principal investigator:
Dr Pek Kei Im
Lead institution:
University of Oxford, Great Britain

Alcohol consumption is a major cause of death and disability worldwide. Despite its hazards, alcohol consumption is still widespread and has been increasing in many low- and middle-income countries. Moreover, the associations of alcohol drinking with many diseases and causal nature of the associations, especially moderate drinking, remain uncertain. Furthermore, the underlying mechanisms through which alcohol influences different diseases are still poorly understood.

The proposed research aims to investigate comprehensively the causal relevance of alcohol drinking for a phenome-wide range of mental and physical health outcomes and to explore the underlying biological mechanisms. It will utilise genetic, multi-omics and extensive health record data in the UK Biobank. There are four integrated work packages, covering (i) associations of alcohol drinking with health-related traits and disease risks; (ii) genetic epidemiology to assess causality of alcohol-disease/traits associations; (iii) multi-omic (proteomics, metabolomics) approaches to identify novel alcohol-associated biomarkers and disease pathways; and (iv) alcohol-attributable disease burden. The findings will be compared and meta-analysed with those from large-scale prospective cohort studies in other diverse populations (e.g. China Kadoorie Biobank).

It is expected that the primary analyses will be complete within 3 years, with applications for extension to allow for secondary analyses and more in-depth investigations. The study findings will expand the understanding of the scope and causal pathways of alcohol-related health impacts, contributing evidence to advance prevention and management of alcohol-related diseases and inform alcohol control policies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-history-and-health-consequences-of-rare-variants

Assessing the history and health consequences of rare variants

Last updated:
ID:
12788
Start date:
1 April 2016
Project status:
Closed
Principal investigator:
Professor Gilean McVean
Lead institution:
University of Oxford, Great Britain

Each individual carries many rare genetic variants (frequency less than 1 in 1000), several of which affect gene function and may affect disease risk. Moreover, rare variants, being typically recent in origin, are often geographically restricted and more so than common variants, which can cause difficulties for genetic association studies. We will analyze the variants represented on the UK Biobank Axiom array to characterize the distribution of rare variants between individuals, estimate the penetrance of disease-causing variants understood to cause severe genetic disease and to infer their evolutionary history (age, geographical origin, recurrence and evidence for purifying selection). By assessing the penetrance of known ?disease-causing? rare variants the research will provide an unbiased assessment of the health consequences of particular types of genetic alteration in individuals not ascertained for a particular disease (or without a family history). Moreover, by assessing the correlation between genetic and geographic proximity (population stratification) we will gain more powerful and better controlled tests for estimating the disease risks associated with rare variants. Finally, by understanding the evolutionary history of variants, we will estimate the mutation and selection pressures associated with different types of genetic change. Rare genetic variants (frequency of less than 1 in 1000) will be identified within each individual from the Axiom array data. The likely impact of such variants on gene function and disease will be inferred by comparison to external databases. The penetrance of mutations will be estimated by analyzing medical data available on cohort participants (e.g. cancer registries, hospital admissions, prescription records). Relatedness structures, evolutionary history, and correlation with geography will be inferred by measuring the sharing of combinations of variants (haplotypes) around rare variants and linking information to data available on participant?s geographic data. Full cohort


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-human-visual-brain-in-the-face-of-eye-disease

Assessing the human visual brain in the face of eye disease

Last updated:
ID:
705478
Start date:
17 April 2025
Project status:
Current
Principal investigator:
Dr Koen Vincent Haak
Lead institution:
Tilburg University, Netherlands

This research project aims to better understand how the brain responds to common eye diseases such as age-related macular degeneration and glaucoma. Previous research has found that altered visual inputs can lead to both neuroplasticity and transneuronal degeneration. It is unclear to what extend these processes vary across individuals and brain structures. Answering this question is important because individual differences in neuroplasticity and transneuronal degeneration can impact visual functioning in daily life and potential routes for treatment and rehabilitation. In this project we will compare visual brain structure and function in ophthalmological patients against normative ranges derived from healthy individuals. This will allow us to derive individualised markers of neural plasticity and degeration in terms of quantifiable deviations from the norm and study the deviation patterns. We will focus on phenotypes that can be derived from the UK Biobank brain imaging data. For instance, we will use the available T1-, diffusion and susceptibility-weighted MRI data to estimate gray- and white-matter properties, as well as iron deposition, alterations of which have previously been linked to neural degenation. We will also assess visual brain function through the analysis of the available resting-state functional MRI data. These functional measures are likely more sensitive to neuroplasticity. Brain structures of interest comprise the visual system, including the retinothalamic and thalamocortical visual white matter tracts, visual cortical areas and the connectivity between them, and the connectivity between the visual cortical areas and cortical areas related to sensory-motor functioning and attention. To ensure rigorous sample selection and patient characterisations, we will also analyse health and retinal imaging data.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-impact-of-early-lifestyle-and-environmental-factors-on-health-outcomes-using-clinical-genetic-and-imaging-data

Assessing the impact of early lifestyle and environmental factors on health outcomes using clinical, genetic, and imaging data

Last updated:
ID:
107031
Start date:
14 March 2024
Project status:
Current
Principal investigator:
Professor Zhichao Jiang
Lead institution:
Sun Yat-Sen University, China

This research project aims to develop new and improved methods for assessing the effects of early lifestyle and environmental factors on health outcomes. These factors, such as exposure to toxins, poor nutrition, and stress, can have long-lasting effects on health, but it is difficult to identify the exact impact due to various issues in observational studies.
The project aims to address common problems in observational studies such as participant dropout, measurement errors, and complex data formats. It will develop methods that use the potential outcomes framework to define causal quantities of interest, establish identification assumptions, propose estimation strategies, and evaluate the sensitivity of the results to the assumptions. These methods will provide more accurate and reliable estimates of the causal effects of early life experiences on health outcomes.
The project will provide a better understanding of how early life experiences can affect health outcomes, which can inform public health policies and interventions. The findings from this research can inform policymakers and healthcare practitioners about the long-lasting effects of early life experiences on health outcomes. This information can help develop targeted interventions aimed at improving the health outcomes of individuals who have been exposed to adverse early life experiences.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-impact-of-environmental-pollutants-on-accelerating-aging-and-identifying-susceptibility-genes

Assessing the Impact of Environmental Pollutants on Accelerating Aging and Identifying Susceptibility Genes

Last updated:
ID:
578096
Start date:
10 March 2025
Project status:
Current
Principal investigator:
Professor Yang Song
Lead institution:
Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, China

Research Questions:
1. Develop and apply aging clock model based on multi-omics, functional performance, cognitive function, and imaging data environmental exposures, and health outcomes to quantify the impact of environmental pollution on biological aging.
2. To identify biomarkers and phenotypic characteristics that could predict accelerated aging due to environmental pollutants.
3. To assess the individual variability in resistance to pollution-induced aging using genomic data and identify potential genetic factors linked to this resistance.
4. COPD is an age-related disease, and both genetics and environment are risk factors for COPD. How do environmental exposures interact with these genetic variants to influence COPD susceptibility?
5. Through a comprehensive GWAS approach, we aim to elucidate the genetic underpinnings of age-related disease and provide insights into the interplay between genetics and environment in disease pathogenesis.
objectives and scientific rationale:
Aging is a complex biological process influenced by genetic, environmental, and lifestyle factors. Environmental pollutants, particularly air pollution, have been implicated in the acceleration of aging, contributing to various age-related diseases.
Aging clocks, which utilize biological markers such as blood biochemistry, gene expression, and omics data to estimate biological age, have become an essential tool for investigating the impact of environmental exposures on aging. By integrating multiple aging clocks with data on environmental exposures, phenotypic age, cognitive function, and functional performance, we can gain insights into how pollutants influence aging across different organs and systems. Moreover, genetic factors likely contribute to individual variability in the response to environmental stressors, making genetic susceptibility a crucial consideration in understanding the mechanisms behind accelerated aging.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-impact-of-natural-environmental-exposures-on-multimorbidity

Assessing the impact of natural environmental exposures on multimorbidity.

Last updated:
ID:
70291
Start date:
14 July 2021
Project status:
Closed
Principal investigator:
Professor Peter Coventry
Lead institution:
University of York, Great Britain

People with multimorbidity have two or more long-term health conditions at the same time. These can either be physical conditions, mental conditions or a combination of both. Multimorbidity is an important topic to study because it has a negative impact on individuals’ lives, healthcare systems and the economy. Those who have multimorbidity are more likely to have poorer quality of life and become disabled. They also tend to take multiple long-term medications to manage their conditions, which have unpleasant side effects. Multimorbid individuals also require complex healthcare management plans and in general tend to use health services more. This puts financial strain on government bodies. However, the natural environment in which people live, work or socialise can have an impact on their health. For example, having greenery (known as green space) or water bodies (known as blue space) in a neighbourhood could improve people’s mental and physical health. This happens in several ways, including more socialisation, increased physical activity and reduction in city air pollution and noise. While, there is research into the ways these natural environments impact a single mental or physical health condition, such as having either depression or diabetes, little research has been done on the impact of having two or more co-existing conditions on a person. This project aims to examine the relationship between different green and blue spaces and multimorbidity in adults. This will involve linking data on the local environment to UK Biobank participants’ residential address. Such an approach will allow us to measure different types of green and blue spaces, such as parks, street trees, lakes or canals. Being able to identify how accessible and available these types of spaces are in the surrounding neighbourhood will further increase our understanding of the ways they might impact multimorbidity differently.

In order to better understand how common multimorbidity is in the population, a range of statistical techniques will be employed. The relationship between different green and blue spaces and the probability of having multimorbidity will then be examined. This research is conducted as part of a PhD project and is expected to take 24 months. It will generate new knowledge and expand the field of environmental health.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-impact-of-past-admixture-and-selection-on-genetic-variants-associated-with-traits-and-diseases-across-diverse-populations

Assessing the Impact of Past Admixture and Selection on Genetic Variants associated with Traits and Diseases across diverse populations

Last updated:
ID:
722971
Start date:
19 September 2025
Project status:
Current
Principal investigator:
Dr Maximilian Larena
Lead institution:
Uppsala University, Sweden

The diversity of human populations has been shaped by past admixture and natural selection, influencing the frequency and distribution of genetic variants linked to traits and diseases. Understanding these processes is key to identifying novel genotype-phenotype associations and their relevance to modern health. Using UK Biobank data, we will develop and assess a streamlined and computationally efficient bioinformatics pipeline to investigate the impact of ancestry specific markers derived from ancient modern human populations (e.g. Hunter-gatherer-related, Farmer-related, etc) and archaic populations (Neanderthal, Denisovan, and unidentified lineages) on genetic variation and its association with specific phenotypes and selection signatures in modern populations. By comparing newly identified variants from our data resource covering underrepresented ancient and present-day African, Eurasian, and Asia-Pacific populations with those in the UK Biobank, we aim to uncover ancestry-specific genetic factors linked to specific diseases and traits. This cross-population analysis will clarify how genetic diversity has been shaped over time and reveal biologically significant variants that may contribute to disease risk. Through advanced computational methods, statistical modelling and evolutionary genomics, our research will provide deeper insights into human adaptation, the genetic architecture of health, and potential applications for precision medicine.

Specifically, we aim to address the following questions:
1) How have ancestry-specific genetic markers from ancient and archaic populations influenced the distribution of disease- and trait-associated variants in modern humans?
2) What are the signatures of recent natural selection on these ancestry-derived variants, and how do they affect health-related traits today?
3) Can integrating ancient and underrepresented population data improve detection and interpretation of genotype-phenotype associations?


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-impact-of-sex-specific-life-events-on-womens-brain-health

Assessing the impact of sex-specific life events on women’s brain health

Last updated:
ID:
103758
Start date:
21 November 2023
Project status:
Current
Principal investigator:
Professor Daniel Bulte
Lead institution:
University of Oxford, Great Britain

Although some brain diseases are known to affect women and men differently (e.g., Alzheimer’s disease), most studies neglect this effect, creating a troubling gap in research data. Some events that might contribute to this discrepancy are pregnancy and menopause, and in fact, these events have been shown to alter the brain, with a significant risk increase for impairment later in life. Nevertheless, there are still very few studies on this topic.
This project will last approximately 3 years and will investigate the health of the female brain. This will be achieved using magnetic resonance imaging (MRI) data as well as cognitive, genetic, demographic information.
The outcomes of this project will have a significant impact not only on understanding the impact of life events on brain health and function, but it will also highlight the critical, but often neglected, aspect of female representation in health research.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-impacts-of-biological-chain-interactions-on-health-and-well-being

Assessing the Impacts of Biological Chain Interactions on Health and Well-being

Last updated:
ID:
705789
Start date:
30 April 2025
Project status:
Current
Principal investigator:
Ms Li Gong
Lead institution:
Southwestern University of Finance and Economics, China

Research question
The human body is a complex network of biological systems, including gene-gene, hormone-hormone, and brain region-brain region interactions, which significantly influence mental and physical health and well-being. Current research indicates that these interactions, manifested through behaviors and emotion, are also affected by environmental factors. Despite advancements, the precise mechanisms of these influences remain unclear, highlighting a gap that this study aims to address.
Objective
This study focuses on understanding how social behaviors and emotional responses mediate the impact of biological interactions and how modifiable environmental factors. We will adopt an integrated and multidisciplinary approach, combining the power of genomics, transcriptomics, proteomics, metabolomics, and comprehensive epidemiological data. By conducting those methods, we strive to uncover novel insights into the biological chain interactions that influence health. Specifically, we aim to identify key molecular pathways, potential biomarkers, and causal relationships that drive the development and progression of various chronic diseases, including but not limited to diabetes, rheumatoid arthritis, and inflammatory bowel disease.
Scientific rationale
Emerging research suggests that interactions among genes, hormones, and brain regions can profoundly affect an individual’s mental and physical health. For instance, the self-esteem and burnout levels of monozygotic and dizygotic twins are influenced by shared genetic factors. The interplay between neuropeptides and cortisol during stressful situations helps regulate stress responses and emotional states. To accomplish this, we will combine multi-omics and epidemiological data to discover novel risk factors, biomarkers and provide definitive evidence for known associations of biological chain interactions and health reported by traditional observational studies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-inflammatory-processes-in-noncommunicable-and-communicable-diseases-including-covid-19

Assessing the inflammatory processes in noncommunicable and communicable diseases, including COVID-19

Last updated:
ID:
64276
Start date:
15 June 2021
Project status:
Current
Principal investigator:
Dr Yachana Kataria
Lead institution:
Boston University (US), United States of America

The COVID-19 pandemic caused by infection with SARS-CoV-2 has led to more than 325,000 deaths worldwide as of mid-May 2020. Although different mechanisms, cardiovascular diseases and COVID-19 share common risk factors, such as smoking, obesity, and type 2 diabetes. Moreover, both diseases are characterized by a disrupted immune response. Disease severity in COVID-19 may be determined by genetic variation in the immune response which could provide insight into potential therapeutic targets. Our research aims to identify pathways which could lead to therapeutic targets in COVID-19 & cardiovascular (i.e. heart disease) diseases.

Currently there is no specific cure or vaccine, and treatments are symptomatic for COVID-19. We aim to determine if genetic variation in the immune system and its association with the presence of one or more disease and its relation to outcome of COVID-19. This will help identify subsets of population that can benefit from these therapeutics.

Similarly, we also aim to determine if genetic variation in the immune system for chronic diseases (i.e. cancer, diabetes, cardiovascular disease etc.) as well. Studies have found genetic variation in the immune system are implicated in cardiovascular disease. However, we don’t know how these genetic variants interact with risk factors such as diet, exercise, sleep, circadian rhythm, alcohol, smoking, and sunlight exposure and how these interact with cardiometabolic risk factors and the risk of disease.

Our study will take advantage of genetic epidemiological study designs. These approaches benefit from understanding the direction of cause and effect relationship. It also takes advantage of genetic variants as proxies for modifiable risk factors to understand the causal relationship.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-influence-of-genetic-and-non-genetic-risk-factors-on-subclinical-and-clinical-cardiovascular-disease

Assessing the influence of genetic- and non-genetic risk factors on subclinical and clinical cardiovascular disease.

Last updated:
ID:
68746
Start date:
11 January 2021
Project status:
Current
Principal investigator:
Professor Georg Ehret
Lead institution:
Geneva University Hospitals, Switzerland

Cardiovascular disease (myocardial infarction, stroke, etc.) is more likely in individuals with a combination of risk factors, such as cholesterol or high blood pressure. In most cases, the genetic contribution to the risk factors originates in the cumulative effects of several altered genes (“many genes – small impact – frequent”). But for a significant proportion of individuals, the genetic predisposition is due to a single change in one gene with large impact, greatly increasing cardiovascular risk (“one gene – large impact – rare”). One prime example for a disease due to a single gene-change is familial hypercholesterolemia (FH). The same logic applies for familial blood pressure elevation (much more rare).
The diagnosis of single rare gene-changes typically requires expensive DNA sequencing (reading the sequence of the DNA letter by letter) to find the abnormal gene.

This study’s primary objective is to design a statistical model that would predict the probability of finding an FH variant by sequencing, based on genetic and non-genetic evidence. Our objective is to prioritize individuals for sequencing.
With the help of data from the UK biobank, we will compute statistical models, including non-genetic data (e.g. cholesterol and blood pressure levels) and genetic data (low-cost genotyping results). We call these models “risk scores” and all participants of UK Biobank will have their individual risk score for monogenic cholesterol and blood pressure elevation. We will then validate these risk scores established in the UK biobank in a smaller sample of the UK Biobank who have sequencing data available and in individuals from Switzerland. We will also analyze the impact of genetic- and non-genetic characteristics on cardiovascular disease, to improve our model and as secondary analyses. We expect the first results by year 2021 and the project is projected to end in 2024.
Our objective is to contribute to much more efficient, cost-effective diagnosis of familial hypercholesterolemia and hypertension by checking patient risk scores before considering sequencing its DNA . This will enable more effective screening of a larger number of cases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-influence-of-modifiable-risk-factors-on-the-occurrence-and-development-of-aging-and-aging-related-diseases-utilizing-multimodal-data

Assessing the Influence of Modifiable Risk Factors on the Occurrence and Development of Aging and Aging-Related Diseases Utilizing Multimodal Data

Last updated:
ID:
470494
Start date:
30 October 2024
Project status:
Current
Principal investigator:
Dr Xiaodong Pan
Lead institution:
Fujian Medical University, China

Aging is a gradual and irreversible pathophysiological process. It presents with declines in tissue and cell functions and significant increases in the risks of various aging-related diseases, including neurodegenerative diseases, metabolic diseases, musculoskeletal diseases, and immune system diseases. With the aging of society, these condition have gradually become the most important causes of disability and death in elderly individuals, which contribute significantly to the global disease burden. Therefore, there is an urgent need to better understand the pathogenesis of aging, identify potential risk factors and develop effective prevention and control strategies.
This project aims to assess how modifiable risk factors and their interaction affect brain health in patients with aging-related diseases. We expect to develop a powerful model capable of accurately assessing an individual’s risk of developing aging-related disorder by utilizing deep learning and integrating various data dimensions, including clinical data, multi-omics data, and exposure factors. In addition, integrating analysis of genetics, proteins, metabolism, and imaging will unveil the regulatory mechanisms of aging-related diseases on brain health, and identify potential new biomarkers and drug treatment targets.
This three-year project will provide valuable insights into valuable information for individual risk assessment and management of modifiable risk factors for aging-related disease, and help to improve brain health and clinical outcomes in patients with aging-related disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-interaction-effects-of-novel-immune-signatures-and-genetic-patterns-across-metabolic-syndrome

Assessing the interaction effects of novel immune signatures and genetic patterns across metabolic syndrome

Last updated:
ID:
147103
Start date:
26 November 2024
Project status:
Current
Principal investigator:
Professor Dong Zhang
Lead institution:
Beijing Chao-Yang Hospital, Capital Medical University., China

Metabolic syndrome have become a major challenge for global public health, which involve the dysregulation of immune cells and molecules. However, the mechanisms of these immune signatures and its interaction with genetic patterns across metabolic syndrome are still unclear. With access to the UK Biobank data including gene sequencing, metabolomics, Olink proteomics and health related outcomes data, we are interested in understanding the relationships between a list of immune signatures and relevance to metabolic syndrome related diseases. In addition, the effects of gene traits correlated with immune cells and molecules will be focused. During 36 months with UK Biobank data, this project will be centered around immune pathway biomarkers, and identify novel immune signatures for potential mechanism across different genetic patterns, thus supporting clinical studies of novel therapy targets for metabolic diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-link-between-physiological-measurements-and-structural-brain-images-using-machine-learning

Assessing the link between physiological measurements and structural brain images using machine learning

Last updated:
ID:
43724
Start date:
22 February 2019
Project status:
Current
Principal investigator:
Professor Galia Avidan
Lead institution:
Ben-Gurion University of the Negev, Israel

The overarching goal of the present proposal is to characterize structural brain changes in healthy individuals in a wide age range. Recent studies attempted to characterize the relationship between brain structure and age by predicting an individual’s age using a structural MRI image. Advancement in the field of machine learning (ML) has enabled remarkable improvement in this domain, with several studies reporting a mean error of ~5 years in predicting age using neuroimaging as input. Linking brain anatomy with age enables to estimate the difference between subjects’ brain-age as predicted by the ML model and the subject’s real biological age or the delta brain age (DBA). We intend to further improve those existing tools, specifically create a more interpretable result, reduce the need for image preprocessing and examine how DBA is influenced by health-related risk or resilience factors. We plan to examine whether genetic markers of rapid or slow aging can be identified. We expect that the project would last 2-3 years, after which the results, the trained model and code would be openly available.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-prevalence-penetrance-and-pathogenicity-of-rare-genomic-variants

Assessing the prevalence, penetrance and pathogenicity of rare genomic variants.

Last updated:
ID:
49847
Start date:
6 April 2020
Project status:
Closed
Principal investigator:
Leigh Jackson
Lead institution:
University of Exeter, Great Britain

Our project aims to use the data from UK Biobank to maximise NHS patient benefit. The majority of research projects that combine DNA and health records are restricted to certain diseases. This project allows us to make new discoveries and test how existing tests and screening approaches would work in a wider UK population. Private companies are selling DNA tests to the public and if this information is not correct, it could cause undue distress, further testing, and potentially lead to unnecessary treatment. We hope to reduce the impact of this by giving more accurate genetic variant interpretation and helping to educate doctors.
We will examine the accuracy of the DNA chip data (similar to those used by these companies) and compare to the higher quality data which examines all of the DNA. By starting with genes which can cause breast and colorectal cancer, we can estimate how many people may be at risk of needless mastectomies, colonoscopies and other interventions if these chips are not very accurate.
We also aim to help patients suffering from rare disease by using the generously donated data from this and other projects to look for new genes or DNA changes which cause disease. We can look across the hospital and GP data for symptoms and diseases and see if groups of people share changes in their DNA which may cause thse. This may lead to new and/or better treatments and more accurate information to help people make decisions about their lives and future children.
Finally, we hope to generate better estimates of lifetime risk and the impact of family history for serious diseases which have a huge impact on people’s lives. We will take everyone with known disease-causing genetic changes in their DNA and see how many end up with that disease and what other factors affect this. Again, this will allow patients to make more informed choices about their care and families. Most current estimates come only from groups who have a family history of disease, we will examine the risk in those who don’t as we suspect the risk to be much lower, potentially saving years of worry for such people who have discovered their DNA results without having a condition first.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-prognostic-and-diagnostic-utility-of-a-sexual-dimorphism-index

Assessing the prognostic and diagnostic utility of a sexual dimorphism index

Last updated:
ID:
533149
Start date:
14 April 2025
Project status:
Current
Principal investigator:
Professor Vincent Detours
Lead institution:
Université Libre de Bruxelles, Belgium

Recent studies have revealed sexual dimorphism (SD) in the transcriptomes of most internal organs. The extent of SD, however, is likely to vary among individuals. We devised an index to measure the degree of SD in specific individuals from any quantitative, possibly multidimensional, phenotype. Our goal is to assess the diagnostic and prognostic utility of our index across disease variables of the UKBiobank (UKBB) and explore its biological basis.

Garg et al. (PMC11390475) showed that many UKBB diseases (ICD10 codes) can be prognosed and/or diagnosed from 67 features including standard blood chemistry and counts and basic anthropometrics. Addition of proteomic features enhanced accuracy. They also demonstrated that their predictors spotted undiagnosed cases, which increased GWAS power. We will evaluate if including our SD index enhances accuracy in this framework.

Our first work package (WP1) will replicate the study of Garg et al., thus use the same fields: 41270, 53, 40000, 40005, 40001, 40002, 40006, 21003, 31, 21842, 21851, 20116, 1558, 20107, 20110, 23165, 22828, 22000, 41271, 41281, 41273, 41283, 41274, 41282, 20002, 20008, 20001, 20006, 20004, 20010, and categories 100006, 100020, 100080, 100083, 100002, 100095, 300, 263.

WP2 will assess if addition of our SD index increases prognostic/diagnostic accuracy and enhance GWAS’ power. In addition to the above fields, sex-related features will be needed (fields 22001, 22014, 22015, and categories 2417, 100068) to compute the SD index and for QC.

WP3 will extend Garg et al. study and WP2 to well-being/health traits in the UKBB physical measures (100006) and online follow-up (100089), e.g. eye measures (100013), hearing tests (100049), cognitive function (100026, 116), and mental health (136).

WP4 will address the biological basis of interindividual SD variation by measuring associations between our SD index and age, circulating sex hormone levels, and genotypes. It requires no additional fields.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-relationship-between-cardiovascular-diseases-and-other-organ-diseases

Assessing the Relationship between Cardiovascular Diseases and Other Organ Diseases

Last updated:
ID:
755281
Start date:
23 July 2025
Project status:
Current
Principal investigator:
Mr Pinyan Huang
Lead institution:
Huazhong University of Science and Technology, China

Cardiovascular disease (CVD) is a leading global health burden and often accompanied by complications associated with the kidney, liver, and brain. The complex interplay between CVD and extra-cardiac organs accelerates disease progression and impairs clinical management, yet the underlying bidirectional mechanisms remain poorly understood. This project aims to understand systemic organ crosstalk in CVD using the UK Biobank’s extensive multimodal dataset.
We will employ a combination of machine learning and causal inference approaches to identify multi-organ interactions. Specifically, we will integrate clinical data, proteomics, and metabolomics to build predictive models for organ-specific complications. Mendelian randomization will be used to assess causal relationships between genetic variants and organ dysfunction outcomes. Models will be validated across age-, sex-, and race-stratified cohorts to ensure robustness and generalizability.
This three-year study is expected to provide mechanistic insights into systemic drivers of CVD-related multiorgan dysfunction and identify high-risk individuals who may benefit from organ-protective interventions. The findings may inform integrated clinical strategies and improve outcomes for patients with complex chronic disease.
We will provide updates on the progress and outcomes of our research project using UK Biobank in annual report. And we plan to disseminate our research findings through publication in peer-reviewed scientific journals.
In accordance with UK Biobank’s AI policy, we will follow prevailing standards for responsible AI when we undertake research using AI. All trained parameters derived from UK Biobank data will be returned as part of our results data submission. We will not use UK Biobank participant-level data in any public generative AI models, nor upload any part of the dataset or derived outputs to public repositories such as GitHub.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-relationship-between-carotid-imt-and-comorbidities-in-their-effect-on-atherosclerosis-development-and-covid-19-related-health-outcomes

Assessing the relationship between carotid IMT and comorbidities in their effect on atherosclerosis development and COVID-19 related health outcomes.

Last updated:
ID:
71257
Start date:
16 March 2021
Project status:
Current
Principal investigator:
Dr Mariya Kuk
Lead institution:
University of East Anglia, Great Britain

Atherosclerosis is a disease which causes thickening and plaque build up within arteries, a type of blood vessels. This may lead to a complete closure of the vessel causing decreased blood flow to the organs affected. Atherosclerosis is thus known to cause heart attacks and/or strokes. Many factors have been identified which may put a person at higher risk of developing this disease. These include increased age, high blood pressure, obesity, diabetes mellitus (body’s inability to properly process the sugar in the blood), as well as abnormal lipids in the blood stream. In addition, new risk factors for this disease development are emerging. These include various bacterial and viral infections, air pollution, other conditions in which the body produces an increased inflammatory response.
Atherosclerosis severity is sometimes measured using a type of imagining called ultrasound, while looking at the arteries in the neck known as the carotid arteries. There, the wall thickness can be measured more accurately, given an estimate on the severity of this disease and prediction of disease progression.
Our project uses the data from the UK BioBank, a database that includes numerous health-related variables from approximately half a million of UK inhabitants, including data such as ultrasound images.
We will be using the carotid vessel thickness in identifying atherosclerosis disease severity and assessing whether other factors may play a role on the atherosclerosis disease development. With the current COVID-19 virus pandemic, we also plan to see whether people who present with a more severe atherosclerotic disease are at a greater risk of infection and worse recovery outcomes when diagnosed with COVID-19, and whether these patients are more likely to have major complications following the infection.

We hope to look whether the following factors may play a role in the development of atherosclerosis:
-exercise
-what the person eats, how often and how much
-any other illnesses including mental health
-social supports from family and friends
-employment and level of education
Based on our research findings, we will develop an equation to help predict patients at the highest risk of atherosclerosis disease development and those who may also be vulnerable to COVID-19. In the future, this might allow health acre providers to identify patients that are at the highest risk of atherosclerosis disease development and poor COVID-19 prognosis (outcomes), and who may subsequently need to be monitored more closely or use better protective equipment to prevent infection.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-relationship-between-chronic-kidney-disease-ckd-and-metabolic-syndrome

Assessing the relationship between chronic kidney disease (CKD) and metabolic syndrome

Last updated:
ID:
489635
Start date:
18 December 2024
Project status:
Current
Principal investigator:
Dr Yang Li
Lead institution:
Zhongshan Hospital Affiliated to Fudan University, China

Chronic kidney disease (CKD) and metabolic syndrome are two major comorbidities affecting a substantial proportion of the general population, with a considerable increase in the associated disability-adjusted life-years lost over the decades. The association between CKD and metabolic syndrome is bidirectional rather than being a cause-effect relationship, as CKD patients are prone to develop metabolic syndrome, and vice versa. Nevertheless, the bidirectional nature and underlying mechanisms of these conditions are poorly investigated. Gaining deeper phenotypic and genetic insights into both diseases could lead to an enhanced understanding of CKD.
Using the UK Biobank, we aim to integrate a wide array of information-encompassing genetic, environmental, lifestyle factors, laboratory indicators, imaging, and omics data-to clarify the epidemiological and phenotypic characteristics of patients with comorbid chronic kidney disease and metabolic disorders. Our goal is to identify potential biomarkers of CKD progression, and temporal order of the CKD and other organ damage. Once identified, we will develop prediction models, early warning systems, and prevention strategies, ultimately helping to reduce the disease burden on both society and families in real-world therapeutic settings.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-relationship-between-clinical-index-metabolomics-life-stylegenomicsproteomics-and-various-types-of-malignant-cancers

Assessing the Relationship between clinical index, Metabolomics, life-style,genomics,proteomics and various types of malignant cancers .

Last updated:
ID:
744724
Start date:
4 August 2025
Project status:
Current
Principal investigator:
Mr Kunlong Li
Lead institution:
Shaanxi Provincial Cancer Hospital, China

Cancer is characterized by uncontrolled cell growth that can affect almost any part of the body . According to recent statistics from the World Health Organization (WHO), cancer was responsible for nearly 10 million deaths in 2020 alone, making it one of the leading causes of death globally. Understanding the epidemiology, etiology, and implications of malignant tumors is crucial for developing effective prevention strategies and improving treatment outcomes.
Objective
To address this problem, the present undertaking aims to employ an integrated approach, leveraging the wealth of genetics, metabolomics, proteomics, and comprehensive epidemiological data. Through multidimensional analyses, we aim to discover new risk factors, identify potential biomarkers, and precise method to find the pathogenesis and treatment of cancer
Scientific rationale
Probing circulating proteins provides unique opportunities to uncover novel biomarkers and improve our understanding of etiology of those diseases. Similarly, blood metabolome is considered as important readouts of aggregated information from genetic factors, gene expression to protein abundance, as well as external environmental factors. To accomplish this, we will combine multi-omics (i.e., proteomics and metabolomics) and epidemiological data to discover novel risk factors, biomarkers and provide definitive evidence for different cancers reported by traditional observational studies. Our endeavor will encompass the analysis of multi-omics and epidemiological information, enabling us to unravel new risk factors, identify potential biomarkers, and comprehend the intricate web of causal relationships of all kinds of cancers.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-relationship-between-flavonoid-rich-dietary-and-hyperlipidemia-following-air-pollution-exposure

Assessing the Relationship between Flavonoid-Rich Dietary and Hyperlipidemia Following Air Pollution Exposure

Last updated:
ID:
887613
Start date:
4 July 2025
Project status:
Current
Principal investigator:
Mr Zhenghong Yao
Lead institution:
Zhejiang Chinese Medical University, China

Research Question
Air pollution is a major global environmental health concern, contributing to cardiometabolic disorders, including hyperlipidemia. Flavonoid-rich foods possess antioxidant and anti-inflammatory properties and may offer protective effects against pollution-induced lipid metabolism disruption. However, population-level evidence supporting this hypothesis remains limited.
Objective
To address this knowledge gap, we aim to utilize the UK Biobank’s large-scale dataset to: Examine the association between long-term air pollution exposure and blood lipid profiles. Evaluate whether higher dietary flavonoid intake mitigates the adverse lipid effects of air pollution exposure. Explore potential effect modifiers (e.g., sex, age, physical activity) in the pollution-diet-lipid relationship.
Scientific Rationale
Ambient air pollutants such as PM2.5 and NO2 may disrupt lipid metabolism through systemic inflammation, oxidative stress, and interference with metabolic pathways. Simultaneously, flavonoids-bioactive compounds abundant in fruits, vegetables, tea, and cocoa-have demonstrated lipid-lowering and antioxidative effects in clinical and experimental studies. However, the interaction between air pollution and dietary flavonoid intake on lipid metabolism at the population level remains poorly understood.
The UK Biobank provides an ideal platform to explore these questions, with its large-scale, prospective design and rich data on environmental exposures, dietary patterns, and lipid biomarkers. We will further incorporate multi-omics datasets (e.g., metabolomics, proteomics) to identify molecular intermediates linking pollution, diet, and lipid outcomes.
We will use multivariable regression, machine learning, and causal inference methods to enhance analytical robustness. This integrative approach will provide novel insights into how diet may modify the adverse cardiometabolic effects of air pollution and inform future preventive strategies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-relationship-between-metabolic-disorders-and-kidney-disease

Assessing the Relationship between Metabolic Disorders and Kidney Disease

Last updated:
ID:
756602
Start date:
3 June 2025
Project status:
Current
Principal investigator:
Dr Xinxin Zhang
Lead institution:
General Hospital, Tianjin Medical University, China

Research question
Kidney disease is a global health problem with high mortality and socioeconomic burden. Metabolic disorders are increasingly recognized as a key driver of the onset and progression of kidney disease. However, reliable metabolic indicators remain lacking for predicting the development of kidney disease.
Objectives
First, we will investigate how metabolic disorders influence the onset and progression of kidney disease, focusing on conditions such as acute kidney injury, chronic kidney disease, diabetic kidney disease, chronic glomerulopathies, and nephrotic syndrome. Next, we aim to elucidate the complex interplay between metabolic disorders and environmental elements, such as lifestyle behaviors (smoking, diet, and physical activity etc.) Lastly, we seek to identify clinically significant metabolic markers and intervention strategies that can facilitate early detection, precise risk stratification, and personalized treatment approaches, ultimately improving patient outcomes and mitigating the global burden of kidney disease.
Scientific rationale
Emerging evidence suggests that metabolic indicators, such as obesity, insulin resistance, diabetes, metabolic syndrome, dyslipidemia and hyperuricemia, play a crucial role in kidney disease progression. However, most existing studies rely on cross-sectional designs or small sample sizes, limiting causal inference and generalizability. Additionally, the interplay between metabolic disorders and environmental factors remains poorly understood. Thus, we will integrate multi-omics approaches (i.e., proteomics and metabolomics) with comprehensive epidemiological data to identify novel metabolic risk factors and biomarkers associated with kidney disease. Our study will analyze large-scale multi-omics datasets alongside clinical and environmental information to uncover new metabolic indicators linked to the onset and progression of kidney disease, providing evidence for early detection and precision medicine.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-relationship-between-obesity-sleep-disorders-and-mental-disorders

Assessing the Relationship between Obesity, Sleep Disorders and Mental Disorders.

Last updated:
ID:
1034472
Start date:
28 September 2025
Project status:
Current
Principal investigator:
Ms Yuyue Yang
Lead institution:
Nanchang University, China

Sleep disorders are common conditions with significant public health implications, and their prevalence has been rising markedly alongside the obesity epidemic. These disorders not only lead to individual health problems such as daytime sleepiness, cognitive impairment, and reduced work efficiency, but also impose a substantial socioeconomic burden. Notably, sleep disorders are often accompanied by multiple comorbidities, with obesity and mental disorders being the most prominent, thereby forming a complex pathophysiological network. These disorders pose substantial challenges for early diagnosis and intervention, as they often exhibit overlapping symptoms and complex neurobiological mechanisms.
Although comorbidity among obesity, sleep disorders, and mental disorders is becoming increasingly common and their associations have been confirmed, the precise causal pathways remain unclear due to the underlying complexity of their bidirectional interactions. This has limited the formulation of effective intervention measures.
To address this gap, the present study aims to adopt an integrated approach by leveraging the rich radiomics, genetics, metabolomics, proteomics, and comprehensive epidemiological data available in the UK Biobank. Through multidimensional analysis, we seek to identify new risk factors, focus on the interplay of the “obesity-sleep disorder-mental disorder” triad, and adopt a multidisciplinary research perspective. We aim to delve into the complex associations between metabolic abnormalities, sleep disorders, and neuropsychiatric symptoms, providing a theoretical basis for the development of integrated treatment strategies. This comprehensive approach will facilitate more personalized and timely interventions, ultimately improving patient outcomes .


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-relationship-between-psoriasis-sodium-intake-and-cardiovascular-disease

Assessing the Relationship between Psoriasis, Sodium Intake, and Cardiovascular Disease

Last updated:
ID:
91687
Start date:
16 September 2022
Project status:
Current
Principal investigator:
Dr Katrina Abuabara
Lead institution:
University of California, San Francisco, United States of America

Rates of psoriasis, an inflammatory skin disease, have been increasing globally, likely due to changing environmental and dietary factors. The disease course and response to new targeted treatments remain highly variable, and there is a critical need to identify modifiable factors that could improve patient outcomes. New research shows that humans may store salt in their skin in response to high levels of salt intake, which could trigger or perpetuate some of the inflammatory processes involved in psoriasis.

The primary aim of our research is to determine whether dietary salt intake is associated with psoriasis. We will also examine whether there are particular subgroups for whom this relationship is strongest. Finally, we will examine whether salt intake helps to explain a previously established relationship between psoriasis and hypertension.

If we find an association between salt intake and psoriasis, future clinical trials may evaluate the utility of low-salt diets, which would be a novel treatment strategy that is low-risk, cost-effective, and widely available.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-relationship-between-sodium-intake-and-il-17-mediated-disease

Assessing the Relationship between Sodium Intake and IL-17-Mediated Disease

Last updated:
ID:
228974
Start date:
18 June 2024
Project status:
Current
Principal investigator:
Dr Katrina Abuabara
Lead institution:
University of California, San Francisco, United States of America

IL-17 is a protein secreted by the immune system that plays a role in multiple inflammatory and autoimmune conditions including psoriasis, psoriatic arthritis, ankylosing spondylitis, hidradenitis suppurativa, systemic lupus erythematosus, rheumatoid arthritis, inflammatory bowel disease, and multiple sclerosis. These conditions are important to study because they are common and confer a large burden on the patients affected by them. Although new treatments targeting IL-17 show great promise, the response to them varies for reasons not fully understood. Studies have shown that sodium plays a significant role in IL-17 activation, therefore, the primary goal of our research is to determine whether dietary sodium (salt) intake is associated with IL-17-mediated conditions. There is evidence to suggest that a history of common infections could influence the role of sodium, therefore we will evaluate whether two common pathogens, Candida albicans and Staphylococcus aureus, impact rates of sodium-mediated IL-17 disease. Finally, because IL-17 mediated diseases are often more common among populations with lower socioeconomic status, we will also assess whether sodium intake and diet quality contribute to this finding. Our results could inform limited dietary guidelines for patients with IL-17-mediated conditions and potentially identify dietary sodium reduction as a low-cost, non-invasive form of disease management.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-relative-contributions-of-environmental-and-genetic-factors-to-cancer-risk

Assessing the Relative Contributions of Environmental and Genetic Factors to Cancer Risk

Last updated:
ID:
807108
Start date:
11 October 2025
Project status:
Current
Principal investigator:
Miss Yushi Jin
Lead institution:
Tsinghua University, China

This study aims to assess the relative contributions of environmental (e.g., smoking, diet, pollution) and genetic (polygenic risk scores, rare variants) factors to cancer risk across tumor types, while testing for gene-environment interactions and identifying high-risk subgroups where genetic predisposition amplifies environmental effects. While GWAS studies suggest genetics explains 5-30% of cancer risk, most cancers are environmentally driven-yet prior studies lack comprehensive exposure data, ignore interactions, and focus on single cancers. Leveraging UK Biobank’s detailed lifestyle, biomarker, and genetic data, we will (1) quantify population-attributable fractions of key risk factors, (2) model statistical interactions, and (3) stratify risk to inform precision prevention strategies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-risk-for-cardiovascular-events-and-falls-after-initiating-antihypertensive-drugs

Assessing the risk for cardiovascular events and falls after initiating antihypertensive drugs

Last updated:
ID:
829202
Start date:
9 July 2025
Project status:
Current
Principal investigator:
Dr Peishan Ning
Lead institution:
Central South University, China

Research Questions:
1. Do different antihypertensive drugs have varying effects on cardiovascular events and fall risk in middle-aged and elderly hypertensive populations?
2. How do these effects differ across subgroups with distinct characteristics?

Objectives:
This study aims to:
1. Compare the impact of different antihypertensive drugs on cardiovascular events and fall risk in middle-aged/elderly patients;
2. Analyze how these effects vary among subgroups (e.g., age, sex);
3. Provide evidence for optimizing treatment guidelines and personalized therapy.

Scientific Rationale:
Antihypertensive therapy reduces cardiovascular morbidity and mortality but may increase fall risk due to side effects such as dizziness and orthostatic hypotension-particularly concerning in older adults, where falls lead to significant injury and healthcare costs. Current research gaps include:
1.No prior study has comprehensively evaluated the trade-offs between cardiovascular protection and fall risk across major antihypertensive classes.
2. Limited data exist on how these trade-offs differ by patient subgroups, hindering tailored treatment strategies.
This study will address these gaps by analyzing real-world and/or trial data to inform precision prescribing and guide tailored treatment strategies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-risk-of-clostridium-difficile-infection-in-inflammatory-bowel-disease-patients-undergoing-various-therapeutic-regimens

Assessing the Risk of Clostridium difficile Infection in Inflammatory Bowel Disease Patients Undergoing Various Therapeutic Regimens

Last updated:
ID:
154360
Start date:
17 April 2024
Project status:
Current
Principal investigator:
Dr Haoran Ke
Lead institution:
Nanfang Hospital, Southern Medical University, China

This research is expected to offer significant value in several key areas, particularly given the public interest in improving the understanding and management of Inflammatory Bowel Disease (IBD) and Clostridium difficile infection (CDI), two conditions that can have a substantial impact on patients’ quality of life.

1. Enhancing Understanding of IBD and CDI Co-occurrence:

By exploring the relationship between IBD, its treatments, and the risk of CDI, this research will contribute to our knowledge of these conditions and their interplay. This is particularly relevant given the increased risk of CDI in IBD patients.

2. Informing Clinical Practice:

The findings could have implications for clinical practice, potentially informing risk assessment, treatment decisions, and patient counseling. If certain treatments for IBD are found to significantly increase the risk of CDI, physicians might opt to modify treatment strategies, particularly in patients at high risk for CDI.

3. Guiding Future Research:

By identifying associations and generating hypotheses, this research could guide future studies, including experimental designs to investigate causality.

4. Public Health Implications:

Given the prevalence of IBD and the serious nature of CDI, the research could have meaningful public health implications. If modifiable risk factors are identified, it may be possible to reduce the incidence of CDI in IBD patients through targeted interventions.

5. Patient Benefit:

Ultimately, the goal of this research is to benefit patients. Enhanced understanding of the link between IBD treatments and CDI could lead to improved management strategies and patient outcomes, reducing the burden of disease and improving quality of life for those living with IBD.

In conclusion, this research aligns with public interest by potentially advancing our understanding of IBD and CDI, informing better clinical and public health strategies, and improving patient outcomes


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-role-of-diet-in-cardiovascular-diseases-type-2-diabetes-and-cancer-a-prospective-cohort-study-and-mendelian-randomization-in-the-uk-biobank

Assessing the role of diet in cardiovascular diseases, type 2 diabetes, and cancer – A prospective cohort study and Mendelian randomization in the UK Biobank

Last updated:
ID:
74221
Start date:
25 January 2022
Project status:
Closed
Principal investigator:
Professor Shiu Lun Ryan Au Yeung
Lead institution:
University of Hong Kong, Hong Kong

What constitutes a healthy diet remains controversial given results differ across studies. The purpose of this study is to explore the role of diet in heart disease, diabetes, and cancer risk in the UK Biobank. We will use different study approaches, in particular genetics, to help generate insights regarding the role of diet in health. The whole project will take 36 months to complete and we expect the findings from this study will be highly relevant to shaping dietary guidelines in different settings.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-role-of-newly-discovered-fruit-and-vegetable-phytonutrients-and-the-impact-on-health-outcomes

Assessing the role of newly discovered fruit and vegetable phytonutrients and the impact on health outcomes

Last updated:
ID:
92495
Start date:
14 September 2022
Project status:
Current
Principal investigator:
Dr Doug Bolster
Lead institution:
Brightseed, Inc, United States of America

Consuming a more plant-based diet has been shown to improve your overall health. We will show that with a detailed, Artificial Intelligence-derived understanding of how plant-based foods (fruits and vegetables) impact our health, we will be able to predict the positive impact of specific fruits and vegetables across a host of health conditions. One outcome of our research will be the development of personalized dietary approaches to address specific health issues faced by an individual. Overall, this project will provide a broader, more flexible, and tailored range of dietary recommendations that will allow individual consumers to maximize the benefits of the foods they eat.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-role-of-notch3-associated-clonal-haematopoiesis-in-vascular-dementia-development

Assessing the role of NOTCH3-associated clonal haematopoiesis in vascular dementia development.

Last updated:
ID:
75265
Start date:
11 October 2021
Project status:
Closed
Principal investigator:
Dr Miguel Ganuza Fernandez
Lead institution:
Queen Mary University of London, Great Britain

Vascular dementia (VD) is the second most frequent type of dementia. About 150,000 patients in the UK currently suffer this degenerative disease. It impacts the ability to perform daily activities. Symptoms include memory loss, slow thinking and difficulty to walk. VD worsens with time. It is caused by the damage of blood vessels in the brain which are not able to bring the oxygen and nutrients that brain cells need, leading to the death of these critical cells. The molecular mechanisms that drive VD are not well understood and there is no cure for VD.

A type of hereditary VD known as Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy (CADASIL) is caused by mutations in a gene known as NOTCH3.

Interestingly, recent data (including ours) indicate that mutations in NOTCH3 that occur over the lifetime of an individual allow mutated blood and epithelial cells to accumulate and expand in healthy individuals.

Our major hypothesis is that the accumulation of NOTCH3 mutated blood cells in healthy individuals can eventually produce symptoms like those present in CADASIL resulting in VD. NOTCH3 mutated blood cells would progressively damage blood vessels by inducing increased inflammation.

Particularly, we think that in some VD patients, blood stem cells (BSC) which produce all our red and white blood cells, have acquired NOTCH3 mutations which made BSCs to divide more or die less than they should. Mutated BSCs accumulate in their bone marrow and blood without initially producing any sign of disease. This accumulation can result in a lot of their blood cells deriving from one single mutated BSC, which it is known as clonal haematopoiesis (CH). CH can be induced by mutations in other genes, especially those driving blood cancer. CH is very common in the elderly and induces a much greater risk of developing blood cancer (11-times higher) and heart diseases (three-fold higher).

This research proposal will investigate if VD patients carry CH with NOTCH3 mutations in their blood by scrutinizing the DNA sequences of VD patients and comparing them with healthy individuals which have not developed VD by the same age. We expect to complete this project within 24 months. Importantly, as there is no known cure for VD, it is mandatory to better understand how this disease arises to provide new therapeutic means for the millions of VD patients worldwide. Towards this, our research will carefully evaluate if CH could drive VD.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-role-of-pharmacogenomics-in-precision-medicine-using-artificial-intelligence-methods

Assessing the role of pharmacogenomics in precision medicine using artificial intelligence methods

Last updated:
ID:
64586
Start date:
15 September 2020
Project status:
Current
Principal investigator:
Dr Georgios Ntritsos
Lead institution:
OpenDNA Ltd, Israel

Recent advances on genome-wide association studies have decipher the genetic architecture of several complex traits and diseases and have identified genes that target already existed drugs. Our study aims to calculate drug specific genetic risk scores in order to access the effectiveness of a therapy that is driven by the genetic signature of the individual.
The proposed research is entirely congruent with the stated aim of UK Biobank to improve the prevention and treatment of a wide range of illnesses. We will examine the effectiveness of drug therapies that will be recommended based on the genetic profile of the individual combined with other demographic and clinical characteristics. These findings will have a major impact to the treatment of the several diseases and the prevention of other co-existing comorbidities.
We will aim for a personalised approach where we will use state-of-the art approaches to calculate a drug-related genetic risk score and we will apply machine learning approaches using the genetic risk score and other clinical characteristics. Through this approach we aim to indicate the best available treatment for an individual when multiple options exist. We expect that our approach will allow for a cost effective treatment response which will be faster and it will reduce the number of the adverse events, the number of prescribed drugs, the number of visits paid to the doctors and days of hospitalisation.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-role-of-rare-and-common-variation-in-human-clinical-conditions-and-risk-prediction-using-improved-methods-for-genotype-imputation-and-genetic-effect-modeling-across-populations

Assessing the role of rare and common variation in human clinical conditions and risk prediction using improved methods for genotype imputation and genetic effect modeling across populations

Last updated:
ID:
84038
Start date:
22 March 2022
Project status:
Current
Principal investigator:
Dr Puya Yazdi
Lead institution:
SelfDecode, United States of America

Many common human diseases are affected by thousands of genetic variants, each with individually small effects on the risk of developing these clinical conditions. Genome-wide association studies (GWAS) identify disease-associated variants by comparing hundreds of thousands of affected and unaffected individuals. The identified variants are then aggregated to produce polygenic risk scores (PRS) to stratify individuals based on their susceptibility to developing a particular disease as a function of the genetic variants they carry. However, the lack of diversity in GWAS which have historically included predominantly those of European descent, and the demonstrated low portability of PRS models built using variants identified in one population to other populations, have limited the utility of PRS in clinical practice. Furthermore, rare variants tend to have a greater contribution to phenotypic variation, but due to challenges in rare variant imputation, are not often included in GWAS.
Over this three-year project, we aim to utilize the data from the UK Biobank (UKBB) to increase polygenic risk score prediction accuracy for its application in personalized medicine. We plan to evaluate and refine conceptually new statistical and computational methods that improve imputation performance of rarer variation, thus driving identification of additional disease-associated variants and improving disease risk estimation. To overcome GWAS challenges associated with non-coding variants, the need for repeated testing for different populations, and to address the massive increase in sample size required to determine if variants are disease-associated, we have developed new exploratory models which identify key disease variants, and novel PRS models which incorporate several levels and types of functional information, and phenotypic and genotypic data.
We will utilize the rich individual-level genetic and phenotypic UKBB data to tackle the influence of lifestyle and environmental factors on human conditions across ethnicities and other socio-demographic groups in addition to the effects of aggregate polygenic risk scores, and evaluate the predictive performance of our novel absolute and relative disease risk models. These models also incorporate biomarkers and other demographic factors, and can be utilized as screening tools to guide health promotion. The results of this project should aid in early identification of individuals at higher risk of certain health conditions who will benefit from preventive interventions and personalized treatments, and will result in the generation of many PRS models for common diseases to optimize clinical care.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-the-role-of-structural-variation-in-human-disease-using-genotype-imputation-with-a-cosmopolitan-reference-panel-of-diverse-variants

Assessing the Role of Structural Variation in Human Disease using Genotype Imputation with a Cosmopolitan Reference Panel of Diverse Variants

Last updated:
ID:
65748
Start date:
6 October 2020
Project status:
Current
Principal investigator:
Dr Michael Zody
Lead institution:
New York Genome Center, United States of America

Genetic variation is a difference in the DNA sequence between individuals. There are many types of variants. The most common affects a single building block of DNA. Another class of genetic variation called structural variants (SVs) can affect tens to millions of consecutive such building blocks. This class of variation accounts for a greater genetic difference between individuals than single sequence changes. Additionally, SVs can have a significant role in disease risk as seen in neurodegenerative disorders, like ALS. Despite their importance, SVs are challenging to analyze and have been understudied. We have created a diverse, high-quality genetic dataset across multiple variant classes, including SVs, that can be used to study the effect of SVs on disease. This dataset has been created from whole-genome sequence (WGS) data on 3,202 samples from 26 populations around the world through an international collaboration known as the 1000 Genomes Project. This dataset can be used as a reference for geneticists looking to study variants that were not directly targeted in their own study, a process called imputation. The idea that a set of variants in an individual of the same ancestry can provide useful information about other variants that were not directly targeted forms the basis of imputation. Imputation increases the likelihood that a genetic analysis will detect an effect (e.g., a gene contributing to the risk of developing a disease) when one truly exists.

Our goal is to use the UK Biobank to study SVs in health and disease. This includes first demonstrating the accuracy of our panel to impute SVs and study the risk these variants have on a disease or trait. For this analysis, we will use height and body mass index (BMI) data. Upon study completion, we will publicly release our reference dataset enabling other researchers to perform SV imputation in their own studies. We will also publish a best-practices guide to SV imputation to promote robust research practices in the community. Next, we will characterize the role of SVs on cardiovascular disease; neuropsychiatric conditions, like autism; and neurodegenerative diseases, such as Alzheimer’s. This analysis may pinpoint genes that contribute to a person’s risk of developing these diseases. We anticipate the duration of this project to be approximately 5 years. Overall, knowledge gained from these studies has the potential to uncover new relationships between SVs and traits, unveil important biological complexities, and ultimately assess individual disease risk.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessing-whether-individual-feedback-results-in-improved-health-outcomes-with-a-focus-on-bmi-feedback

Assessing whether individual feedback results in improved health outcomes, with a focus on BMI feedback.

Last updated:
ID:
9502
Start date:
1 April 2016
Project status:
Closed
Principal investigator:
Dr William Cook
Lead institution:
Manchester Metropolitan University, Great Britain

Research Question: Does informing people that they are overweight
based on their Body Mass Index (BMI) result in individuals improving their health? Does feedback of other health indicators have any effects on individuals?
Outcomes: Weight, BMI, general health.
Unhealthy weight in adults is a growing problem in the UK, with the
Chief Medical Officer recently stating that the problem is in part due to individuals
not recognising that they are overweight. Policy to tackle unhealthy weight in
adults ranges from providing expensive surgery to general public health
information campaigns that are deemed by researchers to be ineffective; the
identification of cost effective interventions to reduce the proportion of people with
unhealthy weight is an urgent issue. The results will have
implications for the efficacy of individualised health feedback as a cost effective
means of delivering public health goals. I will use statistical methods to analyse whether the participant feedback provided is
causally related to improved individual health indicators and whether any such
effects are determined by an individuals? personal characteristics. Full Cohort and the repeat assessment data.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessment-and-characterization-of-protein-protein-interaction-affinity-networks-from-whole-exomes

Assessment and characterization of protein-protein interaction affinity networks from whole exomes

Last updated:
ID:
87272
Start date:
10 November 2022
Project status:
Current
Principal investigator:
Professor Mohammed Nazar AlQuraishi
Lead institution:
Columbia University, United States of America

Proteins are the molecules of action encoded by genetic sequences and dictate a large portion of our biology. These proteins interact continuously in our body to facilitate a myriad of biological functions. We plan on characterizing natural variation in protein interactions in humans to improve our understanding of the normal range of interaction dynamics and therefore help determine when differences arising due to mutations meaningfully deviate from normal biology, and how they impact drug efficacy, disease predisposition, and biological phenotypes. However, measuring the affinity of these proteins to interact is not feasible by experimental means given the large number of protein pairs which can interact in multiple ways and vary between individuals. Recently, computational tools have been developed that take the amino acid sequence (protein building blocks) of proteins to determine their affinity for interacting. This new technology allows us to computationally quantify the interaction affinities of the UK biobank cohort to better our understanding of normal variation which can then be applied to determine meaningful differences in interactions between sub-populations such as those at higher risk of disease onset or adverse clinical outcomes. Given the complicated nature of these protein interactions, traditional statistical and contemporary machine learning approaches will be used to best determine the relationship between interaction network topology and sub-populations. These analytical pipelines would allow for scientists, clinicians, and other healthcare professionals to incorporate a protein interaction perspective into their decision making, ultimately with the goal of improving biological understanding. As this project is computational in nature its duration is dependent on the scope of analytical targets chosen; Targets will consist of biologically and clinically relevant phenotypes of interest such as height, lifespan, and cancer diagnosis. Agnostic of analytical target, determining interaction variation of the population would likely take approximately 2 years to develop an efficient pipeline capable of handling the entirety of the UK biobank cohort. With an additional year taken for the post-hoc analysis to determine meaningful conclusions which can be made along with their ensuing consequences and in some cases experimental followup in cell lines. The inclusion of multiple phenotypic targets for analysis would increase the timeline but would similarly increase the benefit of our work, as such a subset of meaningful targets will be chosen on the basis of prevalence, impact, and pre-existing knowledge.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessment-and-prediction-of-early-neurodegenerative-diseases-and-dementia

Assessment and prediction of early neurodegenerative diseases and dementia

Last updated:
ID:
109607
Start date:
23 October 2024
Project status:
Current
Principal investigator:
Dr Emily Simmonds
Lead institution:
UK Dementia Research Institute, Great Britain

The UK Dementia Research Institute (UK DRI) is a unique MRC institute embedded across six universities with members working seamlessly together as integrated teams to address major questions in Alzheimer’s, Parkinson’s, Huntington’s diseases, FTD/ALS, and Vascular dementia.

Experts across the UK DRI would work collaboratively with the UK Biobank data to create new insights and knowledge which can be applied to treating neurodegenerative disease. For instance, in work over the last year, one team showed that Parkinson’s disease can be predicted, by examining UK Biobank data, 7 years before it would usually be diagnosed.

Some major questions we would examine are:
– What are the earliest changes leading to neurodegenerative disease and dementia?
– What are their causes and how can we disrupt them?
– How do these diseases evolve from being “silent” towards producing symptoms?

The UK DRI has specialist researchers from across the UK working on different aspects of disease using different tools, who will come together to work on the UK Biobank dataset. By examining the genetic data held on participants alongside the biomarkers and MRI images, UK DRI researchers can start to uncover the pathways which are at fault when neurodegenerative disease first start, and to identify ways to treat or prevent the disease.

Project duration: 36 months in the first instance

The impact of this should be to allow investigators to identify neurodegenerative diseases in the general population more readily and with higher accuracy, and much earlier on in life than previously. This should enable testing of preventative treatments administered years before disease symptoms arise.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessment-and-prediction-of-the-impact-of-sleep-regularitygenotype-biomarkers-lifestyle-and-psychosocial-factors-on-brain-health

Assessment and Prediction of the Impact of sleep regularity,genotype, biomarkers, lifestyle, and psychosocial factors on Brain Health

Last updated:
ID:
892891
Start date:
2 October 2025
Project status:
Current
Principal investigator:
Dr Yanxu Zheng
Lead institution:
Third Xiangya Hospital of Central South University, China

The global prevalence of brain health-related disorders continues to rise, posing a severe threat to human health. However, their path remains incompletely understood, and existing treatment options are limited. Early identification of high-risk factors remains a core challenge in public health, particularly in early detection and prevention. Although previous studies have revealed the roles of genetic, and lifestyle factors, the interactive effects and biological mechanisms of these factors require further exploration.This study leverages large-scale databases and integrates prior research findings to conduct multi-stage association analyses, aiming to elucidate the impact of these factors on the onset, progression, and prognosis of neurological disorders. By combining imaging and biomarker data, we will analyze potential mechanisms underlying baseline characteristics across different populations, assess the relative importance of each factor, and provide theoretical support for precision diagnosis and treatment strategies, thereby deepening the understanding of brain health disorders.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessment-of-a-novel-robust-myocardial-strain-metric-for-both-healthy-humans-and-those-after-myocardial-infarction

Assessment of a novel, robust myocardial strain metric for both healthy humans and those after myocardial infarction

Last updated:
ID:
52530
Start date:
31 January 2020
Project status:
Closed
Principal investigator:
Dr Andrew Neil Cookson
Lead institution:
University of Bath, Great Britain

This project aims to use the UK Biobank’s dataset of cardiac magnetic resonance images in order to validate a simple and robust method for quantifying the mechanical function of a patient’s heart following a heart attack.

Strain measures the change in size of the heart throughout the cardiac cycle relative to its size at end-diastole (when the left ventricle contains its largest volume of blood), can be done on a local or global scale relative to the left ventricle. There are strain models based on dividing the heart into 17 segments to find highly localised difference in function, however the results can be difficult to interpret swiftly in the clinical setting and have higher associated errors, and as such strain has not been widely adopted into routine practice. Global strain measures also exist and are more robust, but can obscure localised changes in function. The simplified method proposed here will provide rapid diagnostic information for cardiologists and will help them determine the most appropriate and effective treatment plan for each individual patient.

The proposed method quantifies the degree and rate of mechanical strain in the base, mid-ventricle, and apex regions of the heart. We have shown that it characterises the changes in heart function that occur following a heart attack in an animal experiment, and have found it to be robust and repeatable across different users and medical imaging analysis software. Though other strain methods do already exist, our simple regional approach is more robust and repeatable than these voxel-based methods. Crucially, it is more straightforward to interpret and could be easily tracked over time to monitor the condition of the patient.

With rigorous testing and automation, this metric has the potential to be translated into standard clinical practice, supplementing current methods of determining MI outcome. Using the large datasets available in the UK Biobank we aim to establish normal ranges for these strain and strain rate measures, thus enabling simple guidelines to be produced, which can ultimately be translated to clinical practice.

The project is projected to last between 4 months to 8 months and will form a significant part of my doctoral thesis.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessment-of-disease-progression-and-treatment-effects-in-chronic-diseases

Assessment of disease progression and treatment effects in chronic diseases

Last updated:
ID:
566857
Start date:
10 June 2025
Project status:
Current
Principal investigator:
Dr Jeong Yee
Lead institution:
Sungkyunkwan University, Korea (South)

Chronic diseases, such as cardiovascular diseases, diabetes, respiratory diseases, rheumatic diseases, and cancer, are generally defined as conditions that persist for a year or more and require continuous medical care. As these diseases are long-lasting conditions that typically progress slowly over time and can persist for months, years, or even a lifetime, they often require ongoing management and treatment.
The aim of this study is to explore the relationships between various risk factors and health outcomes in patients with chronic diseases. This will involve examining how demographic, socioeconomic, lifestyle, clinical, and genetic factors influence the progression of chronic diseases, treatment responses, and overall health outcomes.
Participants from the UK Biobank who were aged 40-69 years at the time of recruitment and have been diagnosed with one or more chronic diseases will be included. The outcomes of interest in this study will be focused on two main areas: disease progression, including disease severity and complications, and treatment responses, such as effectiveness and adverse drug outcomes. Demographic factors (e.g., sex, age, race/ethnicity), socioeconomic factors (e.g., income, education level), lifestyle factors (e.g., smoking, alcohol, physical activities) will be collected based on survey data. Health-related outcomes, including algorithmically-defined outcomes and first occurrence data will be used to assess their comorbidities. GP prescription records, including drug name, date of prescription, and quantity will be collected. Lab values, genetic factors, and biomarkers can be considered additional factors that are associated with health outcomes. By identifying and analyzing these factors, the study seeks to uncover potential predictive markers and insights that can help improve patient care, treatment strategies, and long-term health management for individuals living with chronic conditions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessment-of-drug-target-safety-issues-using-human-loss-of-function-variation

Assessment of drug target safety issues using human loss-of-function variation

Last updated:
ID:
96201
Start date:
7 July 2023
Project status:
Current
Principal investigator:
Dr Pengqi Wang
Lead institution:
Tsinghua University, China

In drug development, most compounds that enter clinical trials fail due to efficacy and/or safety issues. Cellular and animal models are useful in the preclinical phases, but they have limited predictive value. In vitro experiments may not address the in vivo conditions, and animal models also face the intraspecies inconsistencies. A well-established model for the efficacy and safety analysis before the clinical trials will largely increase the productivity that the industry needs.

Human loss-of-function genetic variants provide a natural in vivo model to study the effects of gene inhibition. The pLoF carriers and noncarriers can show long-term consequences of reduced protein levels in humans. Sequencing and annotation artefacts are a tricky problem in loss-of-function variant detection, so it is important to carefully filter and curate for high-confidence loss-of-function variants.

Targets of approved drugs are enriched for several ‘privileged’ druggable protein families. G protein-coupled receptors, ion channels, nuclear receptors, and protein kinases account for nearly half of human drug targets. Phosphorylation, catalyzed by protein kinases, is the most important type of regulatory modification in proteins. About 60 protein kinases have been identified as drug targets. But there are more than 500 protein kinases encoded by the human genome, possibly with some potential drug targets. Since protein kinases are involved in various cellular pathways, the effects and safety of potential kinase inhibitors need systematic consideration.

The research aims to assess clinical safety of drug target inhibition using large phenotyping and genomic data. The duration of this research is approximately 3 years. The proposed methods include state-of-the-art annotation and filtering methods for human loss-of-function variation, and statistical approaches to test for associations between any health-related phenotype and gene inactivation.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessment-of-frailty-and-biological-age-using-multi-modal-deep-learning

Assessment of frailty and biological age using multi-modal deep learning

Last updated:
ID:
87802
Start date:
13 July 2022
Project status:
Current
Principal investigator:
Professor Daniel Rueckert
Lead institution:
Imperial College London, Great Britain

Over the next 3 years, we will attempt to develop an artificial intelligence solution that will predict the biological age of a patient’s organs. This model will take into account a subject’s genetic and lifestyle information and analyse the images of the target organ to make its prediction. A large part of the research will be concerned with how best to combine these disparate data sources for maximum performance.

We hold this to be a promising and impactful pursuit due to the effect biological age has on a patient’s disease risk and response to medical interventions. It is crucial for doctors to understand how healthy a patient’s organs are when considering a diagnosis and also before recommending certain interventions due to the variable risk of complications.

We know that lifestyle and genetics are what define our biological state, and thus we expect that combining this information with images of the organ of interest will allow us to make the best and most informed prediction possible. Furthermore, by analysing which parts of the input contributed most to our prediction, we can begin to understand the causal roots of biological-chronological age gaps.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessment-of-genotype-to-phenotype-relationships-for-the-purpose-of-novel-therapeutic-development

Assessment of genotype-to-phenotype relationships for the purpose of novel therapeutic development

Last updated:
ID:
53192
Start date:
17 December 2019
Project status:
Closed
Principal investigator:
Dr Joshua Lichtman
Lead institution:
NGM Biopharmaceuticals, Inc., United States of America

NGM is a research-focused drug discovery company with an 11-year history of making important contributions to basic biomedical research and transforming those discoveries into novel therapeutics. NGM recognizes the immense importance of human genetics and the UK Biobank’s extensive phenotypic data as a means for both novel target discovery and for determining the patients most likely to benefit from the therapies we are currently developing. With clinical programs in the areas of liver disease, metabolic disease, ophthalmology and oncology, the diversity of our interests nicely parallels the diversity of phenotypic data available in the biobank.
NGM would specifically like to explore genotype-phenotype relationships within these data with the ultimate goal of translating those discoveries into new treatments for disease. We will start by applying statistical genetics approaches which will mainly focus on rarely-occurring DNA variants which are found in just a handful of people. By comparing their unique genetics to the broad assortment of traits, we can better understand the relationship between the genes and patient health. Understanding the connection in humans between certain genes and the traits that they govern is an essential first step in developing new therapies that will ultimately benefit a large number of patients. The therapies currently under development at NGM would also benefit as our insight into the human biology of their protein targets is fairly limited. We also plan to publish our findings from this work, as we have done many times in the past (i.e. Hsu et al. Nature 2017, Ge et al. Cell Metab. 2018, Harrison et al. Lancet 2018), so that these data can be shared with the scientific community and the public. This motivation precisely fits the UK Biobank’s desire to support health-related research and advance the public interest.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessment-of-organ-specific-biological-age-based-on-whole-body-mr-data

Assessment of organ-specific biological age based on whole body MR data

Last updated:
ID:
60520
Start date:
22 March 2021
Project status:
Current
Principal investigator:
Dr Sergios Gatidis
Lead institution:
University of Tübingen, Germany

Age is a significant risk factor for cardiovascular, neurodegenerative, oncologic and musculoskeletal disorders and an important patient-associated parameter affecting medical decisions in a clinical context. Due to the high inter-individual variation of age-related morphological and functional changes in individuals, the concept of biological age has been introduced aiming to describe the extent of these age-dependent changes in individuals. Definition of biological age is not clear and mostly relies on genetic, metabolic or functional parameters. Assessment of biological age using medical imaging – although providing potential advantages such as organ-specific BA estimation – however is still not fully investigated mainly due to lack of sufficient amount of standardized cross-sectional imaging data of the underlying population. The UK Biobank MR study provides a standardized data set of thousands of whole body MR data sets together with epidemiological, anthropomorphic, functional and laboratory parameters from the participating individuals.
The aim of this project is to derive organ-specific estimates for biological age based on whole body MRI data and to identify risk factors for accelerate biological aging.
To this end, target organs will be segmented on MR data sets using a deep neural approach. Image-based chronological age estimation will be implemented using a deep learning-based regression model. Subsequently, estimates for organ-specific biological age will be derived in a data-driven iterative approach.
Image-based estimates of biological age will be validated by comparison to biological age estimates from epidemiologic, anthropomorphic, laboratory and functional non-imaging data. Finally, risk-factors influencing biological age distribution will be identified using statistical methods.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessment-of-pleiotropic-disease-associations-of-haplotypes-that-possess-special-features

Assessment of pleiotropic disease associations of haplotypes that possess special features

Last updated:
ID:
44891
Start date:
10 December 2018
Project status:
Current
Principal investigator:
Professor Yutaka Yasui
Lead institution:
St. Jude Children's Research Hospital, United States of America

The aim of this research is to assess whether a specific region of human genome that has certain unusual features is associated with risks of multiple diseases that are seemingly unrelated. In a group of adult survivors of childhood cancer, we have observed such associations. Specifically, we have identified a specific region of human genome with the unusual features and detected that people with different genomic patterns of this region have different levels of risk for many human diseases including cancer, cardiovascular diseases, neurological diseases, and musculoskeletal diseases. Such an association of a single genomic region with multiple (disease) conditions is called “pleiotrophy” in genetics and, if confirmed, it has many important implications on both a) biological/genetic understanding of human genome and its evolution and b) clinical etiology and potential treatment strategies of the associated diseases: both of these are potentially of very high public health impact. However, the size of the childhood cancer survivor cohort was small (~2500) and the associations we could detect were limited by the small sample size. Also, it was a cancer survivor population and whether the observed associations in that special clinical population apply to the general population is unknown and requires further investigation. We, therefore, would like to utilize the much larger UK Biobank population data and examine the associations we previously observed as well as other potential associations that we could not examine with adequate statistical power (Aim 1). We will also extend the analysis to other regions of the human genome that have the similar special features (Aim 2). The expected duration of the proposed study is 24 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessment-of-rare-variants-of-mendelian-genes-in-association-with-traits-and-or-disease-phenotypes-in-uk-biobank

Assessment of rare variants of Mendelian genes in association with traits and/or disease phenotypes in UK Biobank

Last updated:
ID:
41232
Start date:
19 June 2018
Project status:
Current
Principal investigator:
Dr Christopher Randal Bauer
Lead institution:
BioMarin Pharmaceutical, Inc., United States of America

Some diseases are caused by defects in a single gene where you inherit one bad copy of the gene from each parent. We call this pattern of inheritance Mendelian after the geneticist Gregor Mendel, who first described this pattern of inheritance of traits in pea plants. Other diseases have a more complex pattern of inheritance with minor defects in each of many genes contributing to the disease state.

In this study, we will examine whether genes associated with Mendelian diseases might contribute to complex diseases. Some of the rare Mendelian gene mutations found in the cohort can be novel, and we plan to experimentally determine functional impact of novel mutations for a few disease genes we study. This will help us to analyze phenotypic associations with Mendelian genes using only variants that are most likely disease-causing, and it will significantly improve the sensitivity in association analysis. The hope is that if we can find such associations, it might be possible to repurpose existing or develop new drugs for Mendelian disorders to treat more complex diseases.

Our research plan is in agreement with the stated aim of UK Biobank “research intended to improve the prevention, diagnosis and treatment of illness and the promotion of health throughout society”. And we wish to access the full cohort data and carry out our analysis in 3 years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessment-of-the-associations-between-domestic-water-hardness-usage-and-eczema-prevalence-and-incidence-in-middle-aged-adults-aged-from-england-and-wales

Assessment of the associations between domestic water hardness usage and eczema prevalence and incidence in middle-aged adults aged from England and Wales

Last updated:
ID:
70068
Start date:
17 June 2021
Project status:
Closed
Principal investigator:
Mr Diego Jose Lopez
Lead institution:
University of Melbourne, Australia

Eczema common itchy skin condition that develops in children and adults. Hard water (water with a high mineral content) may increase the risk of eczema. To date, studies have only examined the association between hard water and eczema n children. We aim to assess the associations between hard water in people homes and eczema in the UK Biobank study, from the 2006 baseline and 2014 follow-up. Water hardness data will be collected from the Drinking Water Inspectorate and linked to the participant’s home address (in 1-km grids). Associations between water hardness and eczema outcomes will be assessed using regression models while taking into account other factors that lead to eczema and hard water. The project duration will be 14 months to allow for preparing and analysing the data analyses and writing the manuscript for submission to a scientific journal. It is expected that from a better understanding of the effects of domestic water hardness on adult eczema may lead to targeted intervention to decrease eczema incidence, recurrence, and severity. It is expected that from a better understanding of the effects of domestic water hardness on adult eczema may lead to targeted intervention to decrease eczema incidence, recurrence, and severity.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessment-of-the-distribution-of-variants-in-genes-underlying-neuromuscular-diseases

Assessment of the distribution of variants in genes underlying neuromuscular diseases

Last updated:
ID:
99541
Start date:
12 July 2023
Project status:
Current
Principal investigator:
Dr Yeting Zhang
Lead institution:
Sarepta Therapeutics Inc., United States of America

We seek to understand the distribution of variants in genes underlying neuromuscular diseases, including Duchenne Muscular Dystrophy (DMD), resulting from mutations in the DMD gene, and multiple subtypes of Limb-Girdle Muscular Dystrophy (LGMD), resulting from mutations in one of more than thirty genes.
DMD is the most common, severe childhood form of muscular dystrophy. Inheritance follows an X-linked recessive pattern and is caused by mutations in the DMD gene. However, it is not clear if large structural variants and smaller variants in the DMD gene are naturally present in a general population. Interestingly, a less severe form of disease, called Becker Muscular Dystrophy also arises from mutations in the DMD gene; typically, these patients have in-frame mutations that create a shortened, yet functional, dystrophin protein. We seek to investigate DMD genetic variants within the UKBB population to understand which, if any, variants do not lead to clinical presentation of DMD. The identification of these mutation types can help guide the development of therapies to treat this disease.
There are more than thirty different subtypes of LGMD, each corresponding to a different causal gene. Unlike DMD, which typically leads to loss of ambulation around age 12 without interventions and death before age 30, LGMD has a varied disease progression, with disease onset and progression extending into adulthood for some subtypes. It is important to understand the relationship between genetic variation and clinical presentations in both symptomatic and asymptomatic individuals.
We therefore propose to investigate, over a 36-month period, genetic variants, including structural variants (CNVs), single nucleotide variants (SNVs) and smaller insertions and deletions (indels) within the known disease causal genes of DMD and LGMD. In addition, when phenotypes are available for muscular dystrophy patients in UKBB (ICD10 code: G71), we will do genotype and phenotype association analyses. Given the small number of patients with these phenotypes, analyses will likely be limited to descriptive presentation of clinical presentation and disease progression over time.
A deeper understanding of genetic variation in genes causal for neuromuscular disease will advance the development of precision genetic therapies for patients living with these rare diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessment-of-the-heart-brain-liver-axis-and-its-contribution-to-brain-health

Assessment of the heart-brain-liver axis and its contribution to brain health

Last updated:
ID:
137528
Start date:
3 January 2024
Project status:
Current
Principal investigator:
Professor Qi Yang
Lead institution:
Capital Medical University, China

It is now well-established that many chronic diseases, for example, cardiovascular and cerebrovascular diseases, neurocognitive decline, and fatty liver disease tend to co-occur, suggesting potential involvement of multi-organ pathways. While there is extensive evidence that heart, brain, and liver disease outcomes are empirically linked, the underlying mechanisms are not well explored. Numerous studies have been conducted on the heart-brain-liver axis, with a predominant focus on proteomics or microstructure. We aim to explore a macroscopic medium to better understand these multi-organ interactions. Blood vessels connect the brain with other organs and transport neurohumoral mediators across the body. Normal structure and functioning of the vascular system are critical for maintaining the health of each organ. Therefore, we try to investigate the heart-brain-liver axis from the perspective of vascular health. Our objective is to discover imaging markers of the vascular system, enabling us to identify underlying mechanisms that can elucidate the interactions in the heart-brain-liver axis.

UK Biobank provides multi-organ imaging (heart, brain, and liver) acquired and analyzed using standardized approaches of hundreds of thousands of people in the UK, thereby providing an ideal platform for the present study.

Our project is expected to span approximately three years, during which we will focus on the post-processing of large-scale image data, statistical analysis, result analysis, and mechanism discussion from a substantial sample size.

A better understanding of multi-organ interdependence is fundamental to population-level risk stratification and disease prevention since the detection of abnormalities in any of the three organs signals an opportunity for early intervention, which may alter the trajectory of disease progression. Insights from multi-system imaging are expected to have a significant impact on the future prevention of complex diseases and reliably improve quality of life and overall survival.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessment-of-the-heart-brain-liver-axis-in-the-uk-biobank-imaging-study

Assessment of the heart-brain-liver-axis in the UK Biobank Imaging study

Last updated:
ID:
59867
Start date:
15 April 2020
Project status:
Current
Principal investigator:
Professor Qiang Zhang
Lead institution:
University of Oxford, Great Britain

Diseases of the heart, arteries and brain are leading causes of illness and death. Furthermore, 7 out of 10 people with liver disease are not aware that they have a liver condition until their first hospital admission to accident and emergency.

Current statistical methods are too simple, for example, assuming that different organ systems do not interact with each other. In this work we will make use of a very large set of measurements of health and improved statistical methods, to combine data from various organ systems together with general information (such as age, gender etc) and environmental factors. We will explore all possible ways in which these different measurements relate to each other. Current tools that aim to predict the risk of developing diseases are based on only a few measurements of single organs or simple blood tests, and do not work well. So as a second goal, we will test if a more inclusive approach, taking into account all the factors identified by our exploratory work, can lead to improved predictions of disease development. We aim to develop novel and robust combined risk assessment tools that can predict medical conditions more accurate and timely. Our work should assist medical practitioners and society in general by improving health care decision making. Ultimately, we hope to support the UK Biobank’s aim to improve the prevention and diagnosis of diseases worldwide.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessment-of-the-integrated-association-of-potential-determinants-of-physical-activity-by-domain

Assessment of the integrated association of potential determinants of physical activity by domain

Last updated:
ID:
46743
Start date:
13 March 2019
Project status:
Closed
Principal investigator:
Dr Ji-Yeob Choi
Lead institution:
Seoul National University, Korea (South)

Recently, it has been reported that the beneficial effects of physical activity exist regardless of the purpose of the daily activity. However, physical inactivity has increased worldwide; thus, identifying the determinants of daily physical activity is important for improving levels of physical activity. The objectives of this study are to evaluate the integrated associations of the potential determinants of physical activity by domain, including leisure-time, transportation, domestic, and occupational domains. Although several previous studies have evaluated potential determinants, these studies were limited; we will go beyond those previous studies by using physical activity variables in multiple domains, establishing a hypothetical pathway model of potential determinants and assessing the direct, indirect, and total effects of them by using structural equation modeling in longitudinal design. In addition, by comparing our results with findings from the Health EXAminee (HEXA) cohort, one of the largest studies in Asia that includes approximately 170,000 Koreans, we will ensure the validity of the results and/or identify cross-cultural differences. In conclusion, our project will contribute to establishing how determinants of physical activity comprehensively affect physical engagement.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessment-of-the-interactions-and-impact-between-metabolic-diseases-and-inflammatory-skin-disorders

Assessment of the Interactions and Impact Between Metabolic Diseases and Inflammatory Skin Disorders

Last updated:
ID:
599197
Start date:
21 April 2025
Project status:
Current
Principal investigator:
Professor Juan Du
Lead institution:
Huashan Hospital, China

Research Questions
1.Shared Risk Factors: Do metabolic diseases and inflammatory skin disorders share common lifestyle factors (e.g., diet, physical activity) that influence disease onset and progression?
2.Immunological Mechanisms: How do metabolic abnormalities (e.g., lipid metabolism, insulin resistance, oxidative stress) modulate immune responses and contribute to skin inflammation in comorbidity?
3.Disease Interactions and Clinical Outcomes: How does the coexistence of metabolic diseases and skin disorders affect clinical outcomes, including disease severity and treatment efficacy?
4.Biomarker and Target Identification: What are the key biomarkers and therapeutic targets for precision medicine and early diagnosis of metabolic diseases and skin disorders?
Objectives
1.To investigate the shared immune mechanisms between metabolic diseases and inflammatory skin disorders.
2.To assess the impact of shared risk factors on disease development and progression in comorbid patients.
3.To propose targeted clinical strategies for improving outcomes in patients with metabolic diseases and skin disorders.
4.To identify biomarkers and therapeutic targets for precise diagnosis and treatment.
Scientific Rationale
Metabolic diseases (e.g., hypertension, hyperlipidemia, hyperglycemia, hyperuricemia) and inflammatory skin disorders (e.g., psoriasis, eczema) often coexist, suggesting shared molecular mechanisms. Metabolic dysregulation, such as insulin resistance and lipid metabolism disorders, can trigger chronic low-grade inflammation, which contributes to skin inflammation. This research aims to explore these interactions using multi-omics data (genomics, transcriptomics, proteomics), identify key biomarkers and therapeutic targets, and propose strategies for better diagnosis and treatment of comorbid patients.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessment-of-vitamin-d-and-calcium-on-the-risk-of-sarcoidosis-by-mendelian-randomization-approach

Assessment of vitamin D and calcium on the risk of sarcoidosis by Mendelian Randomization approach

Last updated:
ID:
62283
Start date:
26 November 2020
Project status:
Current
Principal investigator:
Dr Natalia Rivera
Lead institution:
Karolinska Institutet, Sweden

Aims: To evaluate the causal relationship by vitamin D and calcium in the development of sarcoidosis and adverse secondary outcomes
Scientific rationale: Metabolic abnormalities, such as low levels of vitamin D and high levels of calcium, often occur in sarcoidosis patients. These may be implicated in disease development or the occurrence of adverse outcomes, such as a chronic course of disease and hypercalcemia. Sarcoidosis is a systemic inflammatory disease of unknown cause. Although the disease can affect any organ, it commonly affects the lung and lymph nodes in 90% of the cases.
In general, genetic investigations are useful methods for identifying human disease biomarkers. In the present study, we intend to investigate the genes and genetic variants associated with vitamin D and calcium levels in the development of sarcoidosis and the occurrence of adverse secondary outcomes, thereby measuring the genetic influence of vitamin D and calcium in sarcoidosis via a Mendelian Randomization approach. Mendelian randomization is a genetic methodology that assesses the effects of gene and/or genetic variants associated with one disease onto another disease.
Project duration: 12 months
Public health impact: The short-term goal is to characterize the genetic effects of vitamin D and calcium in the development of sarcoidosis and the occurrence of adverse secondary outcomes. Indeed, our investigation will enable us to identify sarcoidosis patients with a high risk for adverse outcomes, such as chronic disease course and/or hypercalcemia. The long-term goal is to find substantial scientific evidence that can be translated into clinical applications, such as early public health interventions and health promotion.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assessment-the-impact-of-risk-factors-on-lung-adenocarcinoma

Assessment the impact of risk factors on lung adenocarcinoma

Last updated:
ID:
260281
Start date:
10 September 2025
Project status:
Current
Principal investigator:
Miss Sitong Guo
Lead institution:
National Institute of Biological Sciences, Beijing, China

Lung cancer remains a significant public health challenge, with adenocarcinoma being the most common subtype. However, what factors are risky in lung cancer and how these risk factors influence tumor progression are unclear. The proposed research aims to elucidate the risk factors contributing to lung adenocarcinoma tumorigenesis, investigate their impact on tumor progression, and explore intervention strategies. The project will utilize whole exome sequencing (WES) and whole genome sequencing (WGS) data, along with detailed clinicopathological metadata, to assess the influence of various risk factors on lung cancer survival. Additionally, the study will compare the prevalence of risk factors and their association with survival across different racial and geographical populations. The analysis will involve identifying tumor mutation burden and driver genes, predicting neoantigen load, and inferring human leukocyte antigen (HLA) genotypes. Multivariate Cox regression analysis will be employed to determine the risk factors related to lung cancer survival, considering factors such as smoking, gender, age, geographic regions, ethnicity, and genetic factors. The project aims to stratify patients based on distinct disease biology and identify suitable treatment approaches for more precise management. Overall, this research will provide valuable insights into the complex interplay between genetic and non-genetic factors in lung cancer progression, with potential implications for public health and personalized cancer care.

The findings from this research have the potential to have a significant public health impact by providing insights into the factors driving lung cancer progression and survival disparities across diverse populations. By identifying novel risk factors and developing personalized treatment approaches, the study aims to contribute to improved patient outcomes and inform public health strategies for lung cancer prevention and management.

The project will last 36 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-analysis-between-inflammatory-diseases-and-clonal-hematopoiesis-of-indeterminate-potential-chip

Association analysis between inflammatory diseases and clonal hematopoiesis of indeterminate potential (CHIP)

Last updated:
ID:
86996
Start date:
28 April 2022
Project status:
Current
Principal investigator:
Dr Denny Sun
Lead institution:
Genome Opinion Inc., Korea (South)

Clonal hematopoiesis of indeterminate potential (CHIP), is an age-related phenomenon characterized by a gradual replacement of polyclonal leucocytes by one or more clones marked by somatic mutations. CH is associated with an elevated relative risk of developing hematological malignancies compared to age and sex-matched controls without CH, and an elevated risk of developing non-malignant, immune and inflammatory disorders such as atherosclerotic cardiovascular disease (CVD), Alzheimer’s disease, and COPD.

Our company recently generated high quality and deep coverage NGS data with 1,000x from various diseases and healthy control cohorts. Our aim is to assess the relationship between CH and non-malignant diseases in our dataset and UK Biobank (UKBB), an ongoing, prospective UK cohort study of approximately 500,000 community-dwelling participants aged 40-69 years when recruited between 2006 and 2010.

CHIP affects 10% of the population older than 70 years and has been linked with an increase in cardiovascular and hematological malignancies based on 2% variant allele frequency. However, CHIP somatic mutation calling is very various and limited depending on their data quality and depth of coverage. Recently. a few studies have also reported 1% VAF as a result of statistically clinical significance. Using the UK, we will characterize CHIP and then validate statistical clinical significance for our disease cohorts. Also we will assess for a reasonable VAF cut-off to show statistical significance. Over the next three years, we will use data from UKBB to identify and validate novel disease associations of CHIP as below:

1) Characterize the presence of CHIP in the UKBB. CHIP somatic mutation calling by various cutoff conditions

2) Assess for variant allele frequency distribution between out dataset and UKBB, and limitation for characterize the presence of CHIP

3) Utilize the breadth of available phenotypic data to conduct a comprehensive understanding of the driver mutations (or genes) of CHIP


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-analysis-causal-inference-and-modeling-of-genetic-environmental-lifestyle-and-clinical-determinants-in-the-occurrence-progression-and-prognosis-of-age-related-diseases

Association analysis, causal inference, and modeling of genetic, environmental, lifestyle, and clinical determinants in the occurrence, progression, and prognosis of age-related diseases

Last updated:
ID:
170605
Start date:
10 April 2024
Project status:
Current
Principal investigator:
Dr Fan Zhixing
Lead institution:
China Three Gorges University, China

Diseases associated with aging, such as coronary heart disease, hypertension, heart failure, arrhythmia, diabetes, metabolic syndrome, chronic obstructive pulmonary disease, chronic kidney diseases, stroke and dementia, have become major challenges to global public health, and there is an urgent need to accurately identify their risk factors and develop personalized prevention and control strategies. Previous studies have pointed out that genetic factors, environmental exposure, lifestyle and clinical factors can affect the occurrence, development and prognosis of age-related diseases. However, whether these factors are really related to age-related diseases and how they affect the occurrence and development of diseases still needs large-scale research. Our research aims to analyze the associations of genetic, environmental, lifestyle, and clinical determinants with the occurrence, progression, and prognosis of aging-related diseases, explore their causal relationships, and construct comprehensive models to predict the occurrence, progression, and prognosis of aging-related diseases.
The three-year project contributes to the diagnosis, treatment and prognosis of age-related diseases and to inform public health strategies. The implementation of this project will help to clarify the mechanism of the occurrence and development of age-related diseases, discover the high-risk groups of age-related diseases, realize early prevention and early treatment, so as to reduce the occurrence of age-related diseases and improve their prognosis.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-analysis-of-environmental-exposures-lifestyle-factors-and-noncommunicable-chronic-diseases-a-uk-biobank-cohort-study

Association analysis of environmental exposures, lifestyle factors, and noncommunicable chronic diseases: a UK Biobank cohort study

Last updated:
ID:
916666
Start date:
10 July 2025
Project status:
Current
Principal investigator:
Mr Huan Liu
Lead institution:
Southeast University, China

Research questions
Noncommunicable chronic diseases (NCDs) constitute a growing global health burden, yet the interplay between environmental exposures and lifestyle factors in their pathogenesis remains poorly characterized. Current studies often examine isolated risk factors, failing to address the systematic integration of multi-dimensional exposome data.

Research objectives
This project aims to: (i) Investigate the interactions among air pollution, lifestyle factors, and genetic predisposition in influencing the onset, progression, and mortality risk of NCDs, based on general and genetic data; (ii) Develop novel models for early screening and diagnosis of NCDs using general and multi-omics data; (iii) Further explore potential biological pathways and mechanisms underlying NCDs by integrating omics data and corresponding genetic data.

Scientific rationale for the research
The escalating prevalence and mortality trends of NCDs, driven by demographic aging, rapid urbanization, and lifestyle modifications, impose substantial burdens on healthcare infrastructures and socioeconomic systems. Environmental exposures (notably ambient air pollutants and noise exposure) coupled with lifestyle factors (including tobacco use, suboptimal dietary patterns, physical inactivity, and obesity) serve as principal etiological determinants in NCD pathogenesis. Epidemiological evidence demonstrates significant positive associations between chronic PM2.5 exposure and incident cardiovascular disease as well as pulmonary neoplasms, while sedentary lifestyles and nutritional imbalances have been established as predominant risk factors for type 2 diabetes mellitus and metabolic syndrome. Nevertheless, the synergistic interactions among these exposures and their combined effects on disease trajectories remain incompletely characterized, particularly with respect to gene-environment interplay and long-term exposure dynamics.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-analysis-of-skeletal-health-and-major-chronic-diseases

Association Analysis of Skeletal Health and Major Chronic Diseases

Last updated:
ID:
841746
Start date:
4 September 2025
Project status:
Current
Principal investigator:
Miss Meijun Chen
Lead institution:
Southern Medical University, China

This study aims to investigate the associations between bone health indicators (including osteoporosis, osteoarthritis, and fractures) and chronic comorbidities such as hypertension, diabetes, psychiatric disorders, and sarcopenia. Rather than focusing solely on bone mineral density (BMD), we consider a broader spectrum of skeletal conditions. The objective is to uncover how chronic diseases influence bone health and vice versa, using large-scale population data to support integrated prevention and treatment strategies.

Method: Using data from the UK Biobank, we will conduct both cross-sectional and longitudinal analyses. Logistic regression models will evaluate the co-occurrence of skeletal and chronic diseases, adjusting for variables like age, sex, BMI, and lifestyle. Longitudinally, Cox regression will assess whether baseline bone conditions predict the onset of chronic diseases. Mediation and pathway analyses will be applied to investigate mechanisms involving circulating proteins such as osteocalcin and other metabolic mediators.

Project duration: The duration of this project is approximately 3 years.

Scientific Rationale: Bone is increasingly recognized as an endocrine organ that influences systemic health. Molecules secreted by bone cells, such as osteocalcin, can regulate glucose metabolism, fat storage, and inflammation. Conversely, metabolic disorders like insulin resistance or chronic inflammation can disrupt bone remodeling. Therefore, chronic diseases and bone health likely interact through shared biological pathways that this study seeks to clarify.

Public health impact: Understanding the two-way relationship between bone health and chronic diseases can improve early identification of individuals at dual risk. This could support integrated clinical practices and personalized interventions. Discovering shared molecular pathways may offer therapeutic targets that simultaneously benefit.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-and-potential-biological-mechanisms-of-mental-disorders-and-their-comorbidities-with-aging

Association and potential biological mechanisms of mental disorders and their comorbidities with aging

Last updated:
ID:
452018
Start date:
15 November 2024
Project status:
Current
Principal investigator:
Professor Wei Bai
Lead institution:
Jilin University, China

Mental disorders, as complex conditions influenced by multiple interrelated factors, not only cause substantial distress to individuals but also impose a significant burden on society. Individuals with mental disorders often experience an increased risk of various chronic physical diseases, which further exacerbates their physical and psychological burden. Aging is a normal biological process commonly associated with declines in the function of multiple organs and systems. Although previous research has preliminarily indicated that mental disorders and their comorbidities may accelerate the aging process, these studies have often been limited in scope, and the potential intrinsic connections among these factors remain unclear. This study aims to further elucidate the associations between mental disorders and their comorbidities with aging through a large cohort study, Mendelian randomization analysis, and polygenic risk scores, and to investigate the underlying disease mechanisms.
The project will span three years, with the timeline approximately divided as follows: (1) First year: We will involve topic selection based on research objectives, data cleaning, data organization, and preliminary analysis; (2) Second year: We will focus on validation using additional databases, manuscript writing, and submission; We will also try to explore relevant interesting topics based on preliminary findings; (3) Third year: We will be dedicated to manuscript revisions and reorganization of database content.
This research will enhance understanding and awareness of aging in individuals with mental disorders, assisting policymakers in developing targeted measures for the elderly population, thereby contributing to the goal of global healthy aging. Additionally, by further exploring the causes of accelerated aging, this study will provide new insights and directions for future research on aging, optimize public health policies, and reduce the societal healthcare burden.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-and-prediction-models-for-complex-phenotypes-characterized-by-different-genetic-architecture

Association and prediction models for complex phenotypes characterized by different genetic architecture.

Last updated:
ID:
81202
Start date:
17 January 2022
Project status:
Closed
Principal investigator:
Ms Hannah Klinkhammer
Lead institution:
University Hospital Bonn, Germany

The goal of the proposed project is to develop and apply statistical methods to better predict the individual risk of patients for a specific disease based on genetic data.

Current attempts to predict certain traits or phenotypes such as e.g. diseases or physical characteristics based on individual genomic data lack to detect all of the expected heritability. This might partly be due to the use of simplified approaches to model the association of genotypic and phenotypic data. For example polygenic risk scores (PRS), which measure the individual genetic risk for a certain trait, are usually constructed considering each genomic variant independently and not by analyzing all variants simultaneously.

Finding more accurate models with higher prediction ability can help to tailor treatments for the individual patient and to improve preventive measures. Invasive therapies might be avoided for people with low risk and appropriate preventive measures can be taken for high risk patients. The aim of the project is to develop those integrative and flexible models combining different sources of information. Building such disease risk and stratification algorithms requires a significant amount of resources and effort. Hence, we believe that a duration of 36 months is required to achieve the aims of this project.

The algorithms will be implemented in tools which will be made available as open-source software for potential application in the biomedical field (e.g., disease risk stratification).


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-and-predictive-modeling-of-sleep-cognitive-function-biomarkers-and-mortality-outcomes

Association and predictive modeling of sleep, cognitive function, biomarkers, and mortality outcomes.

Last updated:
ID:
778407
Start date:
27 June 2025
Project status:
Current
Principal investigator:
Dr Le Shi
Lead institution:
Peking University Sixth Hospital, China

1. Research Questions:
This study aims to explore the association between sleep, cognitive function, related biomarkers, and mortality outcomes.
2. Objectives:
Using multidimensional sleep parameters obtained from self-reports, accelerometer measurements, and sleep-related diagnoses from medical records, this study will systematically evaluate the longitudinal relationship between sleep, cognitive decline, and mortality.
3. Scientific Rationale:
3.1 Exposure Variables: 1) Objective sleep parameters, including sleep duration, sleep onset time, sleep efficiency, sleep fragmentation, and calculated rest-activity rhythm parameters, will be obtained from multi-day wrist accelerometer data. 2) Subjective sleep patterns will be assessed using validated sleep questionnaires and self-reports. 3) Sleep disorder diagnoses (ICD codes) and sleep medication usage will be extracted from participants’ medical records.
3.2 Outcome Variables:
Cognitive decline will be evaluated using longitudinal cognitive function test scores. Medical records will provide diagnoses and treatment records related to cognitive impairment and mortality. Additionally, longitudinal neuroimaging data, related fluid biomarkers, polygenic risk scores for cognition, and multidimensional cognitive outcome parameters will be incorporated.
3.3 Analytical Methods:
A sleep-related longitudinal cohort will be established, and cox proportional hazards regression models and survival analyses will be constructed to assess the impact of multidimensional sleep parameters on cognitive decline and mortality events. Sensitivity analyses will be conducted across subgroups, including different age groups, sex, sleep disorder history, family history of dementia, and APOE-4 allele carriers. Additionally, linear regression models will be used to investigate associations between sleep parameters, fluid and imaging biomarkers. Analyses will be conducted using R and Python.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-accelerometer-data-and-circulatory-diseases

Association between accelerometer data and circulatory diseases.

Last updated:
ID:
570174
Start date:
16 June 2025
Project status:
Current
Principal investigator:
Miss Ling Dao
Lead institution:
First Affiliated Hospital of Zhengzhou University, China

I. Research Questions
Is there a significant association between specific patterns of accelerometer-measured physical activity (such as sedentary time, light-intensity activity, moderate-intensity activity, and vigorous-intensity activity) and the incidence of different types of circulatory diseases (e.g., coronary heart disease, stroke, hypertension)?
Can accelerometer data be used to predict the risk of developing circulatory diseases in the future, and if so, what are the most accurate prediction models based on these data?
How do changes in physical activity levels over time, as measured by accelerometers, relate to the progression or regression of existing circulatory diseases?
II. Objectives
To quantify the relationship between different levels and types of physical activity derived from accelerometer data and the occurrence and prevalence of circulatory diseases in a large cohort.
Develop and validate prediction models using accelerometer data and other relevant covariates to identify individuals at high risk of developing circulatory diseases.
Investigate the impact of modifying physical activity patterns, as monitored by accelerometers, on the clinical outcomes and biomarkers associated with circulatory diseases.
III. Scientific Rationale
Physical inactivity is a well-established risk factor for circulatory diseases. However, traditional self-reported measures of physical activity have limitations in accuracy and detail. Accelerometers provide objective and continuous data on physical activity, allowing for a more precise assessment of activity patterns and their potential impact on health.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-acute-pancreatitis-and-subsequent-cerebrovascular-diseases

Association between acute pancreatitis and subsequent cerebrovascular diseases

Last updated:
ID:
98434
Start date:
3 April 2023
Project status:
Current
Principal investigator:
Dr Guoliang Li
Lead institution:
The First Affiliated Hospital of Xi'an Jiaotong University, China

Acute pancreatitis (AP), an acute inflammation of the pancreas, is the most common pancreatic disease, with the incidence of 33.7 cases per 100,000 person-years worldwide and causing a significant health-care burden. The consequences of AP may not only be localized to the pancreas or abdomen but would also be systemic. One of those complications is endocrine dysfunction, and specifically impaired glucose metabolism or diabetes, which could damage blood vessels and nerves. Adults with diabetes are nearly twice as likely to have heart disease or stroke as adults without diabetes.
Cerebrovascular diseases are leading cause of death and disability worldwide. Cerebrovascular diseases are multifactorial with many mechanisms contributing to a complex pathophysiology. Previous studies have identified hypertension, atherosclerosis, cardiovascular diseases and diabetes are the main risk factors contributing to the development of cerebrovascular insult, apart from natural risk factors. One of the major processes worsening disease severity and outcome is inflammation. Cytokines of the IL-1 family are strongly implicated in sterile inflammatory responses that worsen cerebrovascular disease.Effective targeting of the IL-1 system could be therapeutic in the treatment of cerebrovascular disease. Inflammation in AP may spread from the pancreatic vasculature into systemic circulation and promote a generalized inflammatory response, which is deleterious to vulnerable atherosclerotic plaques. Besides, patients with AP have higher rates of cardiovascular diseases. Inflammation, diabetes, and atherosclerosis could be related to the development of AP and cerebrovascular diseases, thereby acting as potential common pathogenic factors.
Our present study aimed to examine the associations of AP with incident stroke in participants from the UK Biobank data set.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-arterial-stiffness-index-and-risk-of-cardiovascular-disease

Association between arterial stiffness index and risk of cardiovascular disease

Last updated:
ID:
3139
Start date:
16 September 2013
Project status:
Closed
Principal investigator:
Professor Paul Elliott
Lead institution:
Imperial College London, Great Britain

Arterial stiffness is an established predictor of adverse cardiovascular outcome but is not currently measured in clinical practice. New devices are developed to facilitate its assessment. For example, the Pulse Trace (Micro Medical) estimates the Stiffness Index (SI) using the volume pulse obtained from an infrared light-transmitting unit placed in the index finger of the subjects. However, the prediction accuracy of the SI has never been evaluated. The goal of the present study is to investigate whether the SI could positively improve our current assessment of patient?s risk of cardiovascular disease.
This study requires access to data from the full UK Biobank cohort and will have three stages. No sample is required. First, we will evaluate the participant?s characteristics and cardio-metabolic markers associated with the SI. Socio-demographic characteristics, anthropometric status, physical activity, diet, biomarkers from blood sample and blood pressure will be analysed. Second, we will investigate whether increased levels of SI can predict the incidence of cardiovascular events, independently of traditional risk factors. We will evaluate whether the prediction accuracy of the SI differs according to the type of cardiovascular disease or according to participant?s characteristics. Finally, we will investigate whether increased levels of SI can predict mortality from cardiovascular disease. Sensitivity analyses will be carried out using the repeat assessment of SI to try correcting the regression dilution bias caused by the measurement error in the baseline SI values.
Cardiovascular disease remains a leading cause of death worldwide. Successful prevention relies on the accurate identification of individuals at risk of developing cardiovascular disease. All the risk prediction models including the traditional risk factors (age, gender, hypertension, dyslipidaemia, smoking, and diabetes) still cannot explain a large proportion of heart disease cases. This study could therefore potentially help to significantly improve the prevention and diagnosis of cardiovascular disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-atrial-fibrillation-and-brain-imaging-characteristics-an-exploration-of-brain-heart-interaction-based-on-uk-biobank

Association Between Atrial Fibrillation and Brain Imaging Characteristics: An Exploration of Brain-Heart Interaction Based on UK Biobank

Last updated:
ID:
763536
Start date:
24 June 2025
Project status:
Current
Principal investigator:
Dr jianhua chen
Lead institution:
University of Goettingen, Germany

Research Questions
What are the structural and functional brain imaging characteristics of patients with atrial fibrillation (AF)?
How do brain imaging features differ between post-stroke patients who develop AF and those who do not?
What are the potential neural mechanisms underlying the increased risk of AF in post-stroke patients?
Can brain imaging biomarkers be used to predict AF occurrence or recurrence?
Objectives
To identify specific structural and functional brain alterations associated with AF using UK Biobank neuroimaging data.
To investigate the differences in brain imaging features between post-stroke patients with and without AF.
To explore potential neural circuits and brain regions involved in AF susceptibility and recurrence.
To develop predictive models for AF occurrence and recurrence based on neuroimaging biomarkers.
Scientific Rationale
Atrial fibrillation is not only a major cardiovascular disorder but also closely linked to brain health. Growing evidence suggests that AF is associated with cognitive decline, stroke risk, and altered brain structure and function. However, the neural mechanisms underlying this association remain unclear.

Using the extensive neuroimaging dataset from UK Biobank, this study aims to elucidate brain imaging characteristics in AF patients and investigate how brain structural and functional changes contribute to AF development and recurrence, particularly after stroke. By identifying neuroimaging biomarkers, this research will improve our understanding of the brain-heart axis in AF and may help in early diagnosis, risk stratification, and targeted interventions for AF patients.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-blood-lipid-fractions-and-cognitive-performance-a-mendelian-randomisation-study

Association between blood lipid fractions and cognitive performance: a Mendelian randomisation study

Last updated:
ID:
30528
Start date:
28 November 2019
Project status:
Closed
Principal investigator:
Dr Zhirong Yang
Lead institution:
University of Cambridge, Great Britain

Observational studies generated inconsistent results regarding the associations of blood lipids and lipid-lowering treatments with cognitive function. There have been no randomised controlled trials primarily investigating the effects of any lipid-lowering therapies on cognitive function. This study involving Mendelian randomisation analysis aims to explore the association of blood lipid fractions with cognitive scores and cognitive impairment in general and stroke population, respectively.
In this study, we will identify British participants who provided data on lipid test results, valid genetic information and cognitive test results. Blood lipid fractions to be analysed in this study include low-density lipoprotein cholesterol(LDL-C), high-density lipoprotein cholesterol(HDL-C) and triglycerides(TG). We are primarily interested in their cognitive test scores for memory, reasoning and information processing speed. We will also investigate the association of blood lipid fractions with cognitive decline. In the Mendelian randomisation(MR) study, we can use genetic variants associated with LDL-C, HDL-C and TG, respectively, to divide participants into subgroups with different genetically determined levels of these blood lipid fractions. While multiple variants are related to each lipid fraction, we will generate allele scores and use the scores as an instrument to evaluate the causal effect of the lipid fractions on cognitive function or cognitive decline. We will compare the results from MR study with conventional observational study.
We expect to spend two years in conducting this study. We will start this project when it has been approved and the data on blood lipid tests are available.
This study will provide further evidence of the unintended effects of lipids and relevant modification drugs on cognitive function in general population and stroke patients. The results of this study may inform lipids management in clinical practice and future research on this topic.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-body-composition-and-measures-of-glycemic-control

Association between body composition and measures of glycemic control

Last updated:
ID:
117233
Start date:
1 December 2023
Project status:
Current
Principal investigator:
Dr Candida Joan Rebello
Lead institution:
Pennington Biomedical Research Center, United States of America

Skeletal muscle is the most abundant tissue in the human body, representing 40% of body weight and contributes to the majority of glucose uptake in response to insulin stimulation. Enhanced muscle mass as observed in body builders is associated with improved blood glucose control and supports the view that enlargement of fat-free mass by training reduces the risk for diseases of impaired glucose metabolism. These results contributed to the notion that fat-free mass is important for maintaining appropriate glucose metabolism. In contrast, some epidemiologic studies show an unexpected positive correlation of fat-free mass with measures of insulin resistance in older men and postmenopausal women. Whether increasing fat-free mass in older adults is beneficial or detrimental for blood glucose control and whether gender differences exist are unanswered questions. The UK biobank offers a wealth of data from gold standard measures of body composition such as dual X-ray absorptiometry (DEXA) and magnetic resonance imaging (MRI) together with blood biomarkers that would provide critical information to answer these questions. We will conduct a population-based retrospective cohort study to determine the relationship between body composition and blood biomarkers. The study will last 36 months and draw from advanced technologies appropriate to the diverse forms of the available data. Impaired glucose metabolism and muscle function predispose to falls, fractures, cognitive decline, hospitalizations, and mortality as people age. This research is consistent with the objective of the UK Biobank to enable greater understanding, prevention, and treatment of a range of serious illnesses.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-bone-mineral-density-and-cardiovascular-disease

Association between bone mineral density and cardiovascular disease

Last updated:
ID:
662558
Start date:
17 May 2025
Project status:
Current
Principal investigator:
Mr Zeng Yupeng
Lead institution:
Jinan University, China

Research questions:
1. Whether the bone mineral density could have an impact on the risk and prognosis of the cardiovascular diseases?
2. Do different bone metabolism indicators associate with cardiovascular diseases?
3. If patients with diabetes will have high risk of loss of bone mineral density?
4. If different dietary and metabolic factors could influence the risk and prognosis of cardiovascular diseases?
Objectives:
1. To investigate the association between bone mineral density and cardiovascular diseases and
2. To detect the link of different levels of bone metabolism indicators with the cardiovascular diseases.
3. To construct a predictive model of the diabetes patients for their potential bone loss.
4. To explore potential relationships between dietary and metabolic factors and cardiovascular diseases.
Scientific rationale:
Based on previous studies, cardiovascular diseases (CVD) referred to variable pathogenesis mechanisms. Metabolic disorders play an important role in CVD, especially in the coronary heart diseases. Conditions such as diabetes and metabolic syndrome have been reported associated significantly with the risk of CVD by many researchers. In addition, plenty of medical clinical and basic research had found that there were potential relationships between bone metabolism and CVD. Given lots of evidence, we want to utilize the unparalleled resource from UK biobank to furtherly investigate the novel features of bone metabolism and other metabolic conditions and to explore their impact on CVD. What’s more, we would also employ the Mendelian Randomization Analysis to find the potential causal relationships.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-brain-damage-and-cardiovascular-cardiometabolic-proteomic-and-genomic-data-in-apparently-healthy-population

Association between brain damage and cardiovascular, cardiometabolic, proteomic and genomic data in apparently healthy population.

Last updated:
ID:
388326
Start date:
19 December 2024
Project status:
Current
Principal investigator:
Professor Karol Adam Kaminski
Lead institution:
Medical University of Bialystok, Poland

Cognitive decline poses a significant risk associated with aging. This decline often accompanies aging and can be exacerbated by several factors such as genetic predisposition, lifestyle choices and underlying health condition.
Neuroimaging is an evolving field that provides insights into the atrophy of anatomical structures and microstructural features of the brain associated with aging. One such feature is white matter hyperintensities, which are non-specific lesions visible on T2 FLAIR images. Their presence increases the risk of dementia and stroke. Other examples of structural damage detectable on MRI scans include impaired water diffusivity and atrophy of the brain and hippocampus. These structural indicators of brain damage are often exacerbated by cardiovascular risk factors such as obesity.

Chronic obesity is a risk factor for cardiovascular diseases and cognitive decline. Advanced biological techniques such as proteomics and metabolomics provide detailed information about the proteome and metabolome, respectively. One well-known biomarker for predicting dementia is amyloid beta protein, associated with Alzheimer’s disease, which can be detected in cerebrospinal fluid (CSF).Since both CSF analysis and imaging tests are not used for regular screening, it is crucial to continue research to understand the mechanisms underlying cognitive decline and to develop non-invasive methods for identifying individuals at risk.

Goal: Understanding the interconnection between cardiometabolic factors, -omics data and asymptomatic brain damage

Hypothesis: preclinical brain damage is related to metabolic disorders, is preventable and might be the beginning of the development of cognitive disorders

Significance:
Understanding associations could offer insights into the mechanisms underlying cognitive impairment in metabolic or cardiovascular disorders and inform targeted interventions for early prevention.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-cancer-therapy-and-cardiovascular-related-comorbidities-in-patients-with-cancer

Association between cancer therapy and cardiovascular-related comorbidities in patients with cancer

Last updated:
ID:
13462
Start date:
1 September 2015
Project status:
Current
Principal investigator:
Dr Nirmala Bhoo Pathy
Lead institution:
University of Malaya, Malaysia

Cancer therapy-induced cardiotoxicity is the leading cause of treatment-associated mortality among cancer survivors. Anticancer therapies may interact with pre-existing cardiovascular risk factors and accentuate cardiovascular morbidity among cancer survivors.
We aim to determine:
1. Prevalence of cardiovascular risk factors in cancer patients
2. Impact of pre-existing cardiovascular risk factors on cancer survival
3. The extent to which cancer therapy, ethnicity, socioeconomic status, and lifestyle, modifies the association between pre-existing cardiovascular risk factors and cancer survival
4. Association between cancer therapy and risk of hypertension, diabetes, stroke and cardiovascular disease among cancer survivors without concurrent comorbidities UKBIOBANK aims to improve the prevention, diagnosis and treatment of life-threatening illnesses including cancer. Knowledge on baseline cardiovascular risk profile and competing mortality risks in cancer patients (cardiovascular mortality versus cancer mortality) will aid tailoring of cancer therapy. In patients with considerable risk of developing cardiovascular disease, physicians may be able to choose adjuvant cancer therapies with higher cardiac safety profile. Furthermore, it will be highly relevant to determine for which patients with cancer will preventive cardiovascular intervention, lead to meaningful reduction in late cardiac complications, and mortality. This research will improve management of cancer patients and their survival. Linkage between UK Biobank and cancer registries allows identification of participants with newly diagnosed cancers.
The baseline presence of cardiovascular risk factors (diabetes, hypertension, hypercholesterolemia, obesity, smoking, and family history of premature coronary heart disease) will be determined in these patients.
Subsequent linkage with death registries will allow estimation of overall and disease-specific survival of cancer survivors. Linkage with primary care data and in-patient hospital admissions data will enable assessment of incidence of hypertension, diabetes, stroke and coronary heart disease in these survivors.
We will compare the survival of cancer patients with cardiovascular risk factors against those without. We intend to include all participants of the UK Biobank cohort with either prevalent, or incident cancers.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-cardiometabolic-health-and-cancer

Association between cardiometabolic health and cancer

Last updated:
ID:
42176
Start date:
16 October 2018
Project status:
Closed
Principal investigator:
Professor Johan Arnlov
Lead institution:
Karolinska Institutet, Sweden

Previous studies have shown that cancer and common non-cancer diseases such as type 2 diabetes and heart disease share several risk factors. In many cases, however, these associations have not been assessed in subgroups of the population, for example in pre- and post-menopausal women. Whether or not shared risk factors play a causal role either in cancer development or cardiovascular disease risk is also largely unknown. We will use genetic and observed data in the UK Biobank to characterise in detail the relationship between heart disease and type 2 diabetes on the one hand, and risk of the most common types of cancer (breast, prostate, gut and lung cancer). The project will take about three years and the results could not only advance our understanding of how cancer develops, but only help to prevent long-term negative cardiovascular consequences of cancer treatment. Fortunately, more people treated for cancer than ever before survive into old age and it is therefore important to identify cancer-related risk factors for cardiovascular and diabetic disease to prevent their occurrence on later life. In conclusion, our project in the intersection between common cancer and non-cancer diseases can benefit persons affected by both disease groups.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-cardiovascular-health-and-non-alcoholic-fatty-liver-disease

Association between cardiovascular health and non-alcoholic fatty liver disease

Last updated:
ID:
96134
Start date:
6 December 2022
Project status:
Current
Principal investigator:
Dr Xiangpeng Ren
Lead institution:
Jiaxing University, China

Non-alcoholic fatty liver disease (NAFLD) has become one of the most prevalent chronic liver diseases and affects 25% general population worldwide. NAFLD is recognized as the liver component of a collection of conditions that are associated with systematic metabolic dysfunction, including abdominal obesity, hypertension, atherogenic dyslipidemia, and insulin resistance, which are also well-established risk factors of cardiovascular disease (CVD). Increasing evidence indicates the presence of NAFLD is associated with an increased prevalence and incidence of CVD.
Recently, the American Heart Association (AHA) proposed a novel measurement of cardiovascular health (CVH), namely Life’s Essential 8 (LE8), to further improve the general population’s health. Given the close associations between NAFLD and the established CVD risk factors, promoting CVH may be an appropriate prevention and management strategy for reducing the burden of NAFLD.
Our study aims to assess the association between CVH and NAFLD in the UK population by conducting a prospective cohort study. Multiple statistical methods will be used to reduce the confounding bias, including multivariable-adjusted regression models and mendelian randomization.
Our study will extend the range of health outcomes associated with the beneficial role of ideal CVH metrics in NAFLD in addition to CVD and indicates that adherence to ideal CVH metrics may be an appropriate prevention and management strategy for reducing the burden of NAFLD as well as other chronic diseases including CVD.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-cerebrovascular-disease-and-parkinson-disease

Association between cerebrovascular disease and Parkinson disease.

Last updated:
ID:
58919
Start date:
2 April 2020
Project status:
Closed
Principal investigator:
Dr Babak Navi
Lead institution:
Weill Cornell Medical College, United States of America

Parkinson disease (PD) is the fastest growing neurological disease currently affecting millions worldwide, yet it lacks an established, commercially-available, imaging-based biomarker that can help identify persons at increased risk. The objective of this research project will be to explore the associations between the burden of cerebrovascular disease on imaging studies and the risk of a subsequent diagnosis of PD. To achieve this aim, we will evaluate the relationship between the degree of vascular changes on brain MRI and carotid ultrasound and the risk for developing PD. Our approach will be a cohort study using longitudinal data from the subset of participants enrolled in the prospective, publicly-available, population-based, UK Biobank study with advanced imaging. The rationale for this research study is that identification of imaging-based risk biomarkers for PD would provide novel insight into the pathophysiology of PD and would enable targeted prevention strategies for those at highest risk. As PD prevalence is expected to double by 2040, it is imperative that actionable risk markers for PD are identified and targeted in order to reduce its rising morbidity. The estimated project duration is 36 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-citrus-consumption-and-skin-cancer-an-analysis-of-risk-and-nutrient-gene-interaction

Association between citrus consumption and skin cancer: an analysis of risk and nutrient-gene interaction

Last updated:
ID:
49419
Start date:
12 July 2019
Project status:
Current
Principal investigator:
Mr Andrew Raymond Marley
Lead institution:
Indiana University Purdue University Indianapolis, United States of America

In the United States, rates of melanoma and NMSC have increased substantially over the past several decades. This increase in incidence has been particularly considerable among fair-skinned/Caucasian populations. NMSC is the most common malignancy in the United States, with an estimated 5.4 million cases and 18,000 deaths annually. Melanoma, the deadliest form of skin cancer, is now the 5th and 6th most common cancer among American men and women, respectively.

Although exposure to UV radiation remains the primary risk factor for skin cancer, recent studies have demonstrated that higher citrus consumption is associated with an increased risk of melanoma and NMSC. Among individual citrus fruits studied, grapefruit consumption was associated with the highest skin cancer risk. Although in need of further confirmation, these findings are reasonable to believe. Citrus fruits naturally contain compounds called psoralens, a type of furocoumarin that is known to increase risk of skin cancer.

Other factors, such as certain physical or genetic characteristics, can also influence susceptibility to skin cancer. We know that people with lighter skin, fairer hair, who spend a lot of time outdoors, or who burn easily are more likely to get skin cancer. However, we do not know if a high consumption of citrus fruits would further increase skin cancer risk in these people. Also, while we do know that certain genes can contribute to skin cancer risk, we do not know if people with these genes, who also have a high citrus consumption, are more likely to develop skin cancer than people who just have the genetic predisposition alone. We also know very little about the genetics behind citrus metabolism and/or what genes may be involved with the increased risk of skin cancer associated with citrus.

Because of that, it is my goal over the next 18 months to: investigate whether high consumption of citrus is associated with melanoma and NMSC in this data set; to analyze whether other skin cancer risk factors influence risk from citrus; and search for genetic markers that may influence skin cancer risk from citrus.

If our assumptions are correct, this research will provide the public with a simple, practical, cost-effective, non-burdensome strategy for reducing skin cancer risk. This research will also increase our understanding of citrus metabolism and how genetics may play a role in this association.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-consumption-of-ultra-processed-foods-and-parkinsons-disease

Association between consumption of ultra-processed foods and Parkinson’s disease

Last updated:
ID:
101148
Start date:
30 June 2023
Project status:
Current
Principal investigator:
Dr Shotaro Haji
Lead institution:
Tokushima University, Japan

Neurological diseases are now the leading cause of disability in the world. Of these, Parkinson’s disease (PD) is the fastest growing, surpassing Alzheimer’s disease. Between 1990 and 2015, the prevalence of PD more than doubled, and disability and death attributable to PD have also increased. The Global Burden of Disease Study currently estimates that 6.2 million people currently have PD. Because patients with PD had lower activities of daily living and quality of life, patients will require care as symptoms progress. Because the incidence of PD increases sharply with age and because the world’s population is aging, the number of individuals affected and caregivers are exposed for exponential growth which is called “Parkinson pandemic”. Thus, there is an urgent need to elucidate the pathogenesis of PD and development treatment methods. However, the pathogenesis of PD remains unclear, but several lines of evidence have indicated the involvement of central and peripheral inflammation in pathological processes. Furthermore, there is the idea, based on Braak hypothesis, that Lewy pathology, the pathological hallmark of PD, begins in enteric nervous system and spreads to brainstem in a retrograde manner, which highlights the importance of the colon as the place of aggregates of Lewy bodies. Inflammatory bowel disease was demonstrated to be a risk factor of PD in a previous meta-analysis and shared genetic effects of LRRK2 between Crohn’s disease and PD risk. These findings suggested important relationship of colonic inflammation resulting altered gut microbiota to PD.
Ultra-processed foods (UPF; e.g., soft drinks, flavorful snacks) have received a lot of attention. They are highly palatable, have a long shelf-life and relatively inexpensive, and can be consumed anytime, and anywhere. However, they are not nutritionally superior. The consumption of UPF is high in many high income countries. For example, the percentage of energy intake from UPF is 29.1% in France, 42% in Australia and 57.9% in the USA. High consumption of UPF can change the gut microbiota and lead to inflammation. Based on accumulating evidence suggested the links between colonic inflammation and PD, consumption of UPF may be associated with onset and development of PD. If we understand the link between UPF consumption and PD, dietary interventions may prevent or suppress the onset or development of PD.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-diabetes-and-risk-of-chronic-kidney-diseases-among-middle-aged-and-older-adults-a-mediation-analysis-on-a-retrospective-cohort

Association between diabetes and risk of chronic kidney diseases among middle-aged and older adults: a mediation analysis on a retrospective cohort

Last updated:
ID:
293286
Start date:
2 September 2025
Project status:
Current
Principal investigator:
Dr Anna Wang
Lead institution:
Monash University, Australia

As far as we know, the present study is the first to explore the association between diabetes and the risk of CKD subtypes. The strengths of our study included its, large sample size, and long follow-up time. Moreover, the data collection was conducted by well-trained technicians with standardized protocols. Second, we used detailed diabetes information to analyze the impact of diabetes on the risk of CKD from multiple perspectives, including the source of diabetes diagnosis. Third, we conducted a mediation analyses, which helped to identify the potential mediating effect between diabetes and the risk of CKD. Our research outcome will support the clinicians to better manage the diatetes to avoid the high prevelence of CKD among China, UK and USA.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-diet-and-depression

Association between diet and depression

Last updated:
ID:
1602
Start date:
1 January 2014
Project status:
Current
Principal investigator:
Dr Gabriella Juhasz
Lead institution:
Semmelweis University, Hungary

Depressive illness is common and costly to the individual and society. Genetic makeup accounts for about 1/3rd of the risk of depression and environmental factors for about 2/3rds. Psychosocial adversity and stress are important aspects of the environment that contribute to depression. Other potentially important environmental factors have been little studied. It is known that what we eat and drink, our diet (carbohydrates, fats etc.) and nutrients (e.g. vitamins) is an important influence on the risk of medical disorders such as obesity, diabetes and cardiovascular disease. These disorders are associated with an increased risk of subsequent depression and are more common in those with previous depression. This suggests that obesity-related disorders and depression may have some similar pathways of risk. The proposed cross-sectional study aims to identify shared and specific interactions between diet and psychosocial and genetic factors for self-reported depression and related disorders. We will use the unique combination of psychosocial, dietary and mental health data available in a subset of 122,000 of the UK Biobank cohort to decisively determine whether or not there are dietary patterns and constituents that lower the risk of self-reported lifetime depression in the face of life stresses. We will then factor-in genetic information (genotyping data will be requested, i.e. no DNA samples) and, using sophisticated statistical techniques, find new dietary and genetic factors that are highlighted because they converge on shared biochemical pathways. Understanding the role of diet in depression meets the UK Biobank?s stated purpose of improved disease prevention; in contrast to genetic and psychosocial factors, dietary behaviour is potentially modifiable. For example, preventative, public health strategies could reduce the prevalence of depression by promoting resilience to psychosocial adversity and by offsetting the biochemical consequences of genetic risk.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-dietary-intake-and-lung-function

Association between dietary intake and lung function

Last updated:
ID:
1163
Start date:
18 September 2013
Project status:
Current
Principal investigator:
Dr Martijn Spruit
Lead institution:
CIRO+, Netherlands

Cigarette smoke is an important risk factor for the development of chronic obstructive pulmonary disease (COPD). However, research suggests that diet may also be important, with some studies finding processed meat and refined grains to increase risk and intake of fruit and vegetables to reduce risk. Until now, it is not clear if the nutrient intake in patients with COPD differs from healthy elderly. Furthermore, it is not known whether diet is related with the severity of COPD. This project aims to investigate 1) the nutrient intake of subjects with COPD in comparison with elderly without COPD and comparable age and 2) the independent association between dietary intake and lung function and lung function impairment, using spirometry measures at baseline. As a secudary objective, we intend to investigate the relation between nutrient intake and incident lunf disease when there are sufficient cases for analyses. This project will help in the understanding of potentially modificable determinants of loss of lung function and severity of obstruction in a British population.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-dietary-intake-and-physical-activity-with-recovery-outcomes-in-stroke-survivors-insights-from-the-uk-biobank

Association between Dietary Intake and Physical Activity with Recovery Outcomes in Stroke Survivors: Insights from the UK Biobank

Last updated:
ID:
785423
Start date:
23 June 2025
Project status:
Current
Principal investigator:
Dr Sanjoy Kumar Deb
Lead institution:
Anglia Ruskin University, Great Britain

Stroke is a leading cause of death and disability globally, with over 1.3 million stroke survivors currently living in the UK. While acute stroke care has improved, survivors often face long-term challenges including recurrent stroke, cognitive decline, depression, anxiety, fatigue, malnutrition, and sarcopenia. These complications can significantly impact quality of life and increase demands on carers and healthcare services.
There is growing interest in the role of diet and physical activity in supporting recovery and reducing the risk of secondary complications following stroke. However, current clinical guidelines for stroke survivors provide limited detail on specific dietary or activity recommendations. Robust, stroke-specific evidence is needed to support more tailored and effective guidance.

Research questions:

Are specific dietary patterns or diet quality associated with the risk of recurrent stroke in stroke survivors?
How are dietary patterns and diet quality associated with cognitive function and cognitive decline over time in individuals with a history of stroke?
Are dietary patterns and diet quality associated with the presence of post-stroke depression, anxiety, and fatigue?
Is diet quality associated with the risk of sarcopenia and malnutrition in stroke survivors?
Does physical activity independently or jointly with diet influence the risk of recurrent stroke, cognitive decline, mental health symptoms, and sarcopenia in stroke survivors?


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-dietary-patterns-bone-health-cardiovascular-disease-and-mortality

Association between dietary patterns, bone health, cardiovascular disease and mortality

Last updated:
ID:
80610
Start date:
30 November 2021
Project status:
Current
Principal investigator:
Professor Tang Liu
Lead institution:
Second Xiangya Hospital of Central South University, China

Nutrition is a significant factor affecting bone health and cardiovascular disease, and it is unclear the potential benefits of nutrition differ according to different dietary patterns on bone health and cardiovascular disease. We would like to perform this study to reveal the role of dietary patterns in bone health and cardiovascular disease to inform health promotion strategies by diet advice. UK Biobank is a prospective cohort study of half a million men and women recruited between 2006 and 2010, and provides the opportunity to investigate prominent hypotheses related to diet and bone health and cardiovascular disease in a contemporary population-based cohort in the UK. It has been previously shown that the dietary data collected from the UK Biobank short food-frequency touchscreen questionnaire, which generally asks about frequency of consumption of main foods and food groups, is highly reproducible. These data are available on all UK Biobank participants. In addition, dietary intakes were re-measured in a large sub-sample of participants (n!=!175!402) who completed at least one online 24-hour dietary assessment and these data can be used to correct for regression dilution and other forms of measurement error. Previous research has commonly reported linear associations of single macronutrients with health outcomes. Our study would like to investigate non-linear associations, adjusted the analysis for intake of macronutrients that could influence the observed associations, and implemented isocaloric replacements based on non-linear and linear associations of nutrients and bone health and cardiovascular disease. The entire project lasts approximately 3 years and it’s helpful for understand the combined and interactive effects of different components of the diet on bone health and cardiovascular disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-early-life-stress-and-psycho-cardio-metabolic-multi-morbidity-the-earlycause-h2020-project

Association between Early-Life-Stress and Psycho-Cardio-Metabolic Multi-Morbidity: The EarlyCause H2020 Project

Last updated:
ID:
65769
Start date:
30 March 2021
Project status:
Current
Principal investigator:
Dr Karim Lekadir
Lead institution:
University of Barcelona, Spain

Early-life stress can be defined as the perinatal or childhood experience of adverse events. It is a risk factor for mental and physical diseases later in life. Since depression, cardiovascular diseases, and metabolic diseases have been found to occur together, one may suspect them to have common behavioural and biological mechanisms. Although the relationship between early-life stress and various individual mental and physical diseases has been previously established, the relationship between early-life stress and the co-occurrence of depression, coronary heart disease, and type 2 diabetes has not yet been addressed. Since the co-occurrence of these diseases implies a heavier health burden for individuals and societies, research on precursors of this co-occurrence is crucial. Therefore, this research project seeks to develop a better understanding of the way early-life stress is associated with the co-occurrence of psychopathology, cardiac disease, and metabolic disease.
Specifically, EarlyCause is a European research project of 14 participating institutions, which aims to evaluate the relationship between early-life stress and the co-occurrence of depression, coronary heart disease, and type 2 diabetes (www.earlycause.eu). Potential common biological mechanisms between early-life stress and the co-occurrence of diseases (e.g. inflammation, neuroendocrine) will be evaluated as well as potential modifying factors (e.g. lifestyle factors). The findings will provide a better understanding of the way early-life stress influences adult health, thereby enabling the identification of risk groups and setting the stage for the development of early interventions to buffer the potential long-term negative health consequences.
We expect this project to take 36 months and the findings to impact public health policies on a global scale. If childhood maltreatment is found to be associated with the adult co-occurrence of diseases, policy makers will be able to use the findings to develop early interventions promoting lifestyles that prevent the onset of the diseases. Avoiding an escalation of adult pathologies among childhood maltreatment survivors will save individuals suffering and societies costs.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-early-repolarization-and-cardiovascular-outcomes-and-metabolic-outcomes

Association between early repolarization and cardiovascular outcomes and metabolic outcomes

Last updated:
ID:
121022
Start date:
27 September 2023
Project status:
Current
Principal investigator:
Dr Yun-Jiu Cheng
Lead institution:
Sun Yat-Sen University, China

Early repolarization pattern (ERP) is a common electrocardiographic variant manifested as elevation of Q wave-R wave-S wave(QRS) junction (J-point) and QRS notching or slurring in multiple leads. Although it has long been considered a benign phenomenon, more recent studies have demonstrated positive association between an ERP and various end points, including ventricular fibrillation (VF) and sudden cardiac death (SCD). We aim to assess the long term prognosis associated with ERP in middle aged and elderly population, in order to help guide clinical prevention decisions. This research project will last for 3 years. The result would prove that early repolarization might be promising a marker of cardiovascular risk in the general population, and repeated measurements of the early repolarization should be implemented in general population to assist in risk stratification and assessment.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-exposure-to-air-pollution-and-acceleration-of-cognitive-decline-in-adults-with-parkinsons-disease

Association between exposure to air pollution and acceleration of cognitive decline in adults with Parkinson’s Disease

Last updated:
ID:
93173
Start date:
26 May 2023
Project status:
Current
Principal investigator:
Dr Jianghong Liu
Lead institution:
University of Pennsylvania, United States of America

The aim of this project is to investigate whether air exposure would accelerate cognitive decline/dementia in individuals who have Parkinson’s disease. This will be done with data from the UK Biobank.
Parkinson’s disease is a progressive neurodegenerative disorder that affects the central nervous system of the brain. Environmental exposures have become an important field of study in epidemiology, and many studies have shown associations between environmental exposures and cognitive deficits. From previous studies, there has been evidence that air pollutants like PM2.5, PM10, NO2 are positively associated with the development of Parkinson’s. We are interested in another facet of this research, specifically how these air pollutants might correlate with cognitive outcomes. Cognitive assessments are an important tool for studying brain development and health. Air pollution exposure has been shown to increase the risk of cognitive impairment and significantly worse performance on cognitive functions like memory and attention. Other studies have shown that those people diagnosed with Parkinson’s also have a much higher chance of having declines in cognition. However, the purpose of our study is to understand the connection between all these areas of study, and see that within the specific population of those diagnosed with Parkinson’s, whether air pollution exposures are associated with accelerated cognitive decline and/or incident dementia.
Our project is estimated to take place over the course of a 12-18 month period once the data is obtained. In this timeframe, we will be collecting data from the UK Biobank on Parkinson’s rates, the outcomes of cognitive assessments/tasks and rates of incident dementia, as well as residential addresses and air pollutant levels. Analysis and interpretation will be performed on these data sets. Results will be presented as a manuscript for publication and conference presentation. We believe that our study will provide a potentially high public health impact and will be an important contribution to the scientific community. Parkinson’s disease (PD) is among the most common neurodegenerative disorders and affects about 10 million people around the world. Similarly, around 50 million people around the world are living with dementia. Findings from this project will help us understand the role of air pollution in development of dementia in Parkinson’s patients. While rates of air pollution exposures increase around the world, it is incredibly important in the public health and epidemiology field to understand the mechanisms of this environmental exposure and how it impacts human brain health.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-factors-influencing-the-life-course-and-the-occurrence-and-progression-of-chronic-noncommunicable-diseases

Association between factors influencing the life course and the occurrence and progression of chronic noncommunicable diseases

Last updated:
ID:
471488
Start date:
10 January 2025
Project status:
Current
Principal investigator:
Professor Yuxia Ma
Lead institution:
Hebei Medical University, China

Chronic noncommunicable diseases (CNCD), including cardiovascular diseases (CVD), nutritional metabolic diseases, cancers, mental and psychological disorders, chronic respiratory diseases and connective tissue, and rheumatic diseases, account for 74% of global deaths, constituting a major public health challenge. An estimated 17 million people below 70!years of age die annually due to CNCD, and 86% of the premature deaths are happening in the middle- and lower-income countries. Therefore, a more comprehensive assessment of the association between the influencing factors and CNCD have significant public health implications. Our study aim to specifically assess the interplay and association between the influencing factors such as genetic factors, age, gender, lifestyle, environment, society, mental health and CNCD, and provide a more effective and comprehensive strategy to prevent CNCD. In our research, multivariable linear regression, logistic regression and cox proportional hazards models will be used to explore the associations between behavioral, mental or social factors and the outcome and mortality of CNCD. Moreover, the potential nonlinear relationships between measurable determinants and CNCD outcomes and mortality will be examined using restricted cubic spline regression. For multiple exposure analysis of environmental determinants, we will use the least absolute shrinkage and selection operator regression, weighted quantile sum regression and Bayesian Kernel Machine Regression. The effects of genetic variants on the above diseases will be investigated by Mendelian randomization. This research can help governments, enterprises, and medical institutions to better understand the factors influencing CNCD, and effectively control the growth of medical expenses to reduce the financial burden on the government, the work burden on hospitals, and the disease burden on patients. It has beneficial effects on the primary and secondary prevention of multiple CNCD.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-food-allergies-and-single-nucleotide-polymorphisms

Association between food allergies and single nucleotide polymorphisms

Last updated:
ID:
43559
Start date:
4 September 2020
Project status:
Current
Principal investigator:
Mr Zeping Shao
Lead institution:
University of Queensland, Australia

Previous researches from our group have noticed that food-allergic patients demonstrating different dietary habits compared with the general non-allergic population. In addition, single nucleotide polymorphisms have been associated with some diet habits. There seems to be a direct link between genetic variants and food allergies. The aim of the proposed research is to characterise single nucleotide polymorphisms (SNPs) associated with food allergies via a genome-wide association study. Moreover, the study will aim at identifying additional associations involving dietary habits, BMI, age, gender, and ethnicity as co-variants. We will also determine interactions between genetic factors, the food allergy status, and the co-variants.
The estimated duration of the current project is 12 months. Results from this study will increase our understanding of food allergies and will emphasize the important role of SNPs/biomarkers. We aim to explore the mechanisms to be identified as therapeutic targets and help identify individuals at high risk for food allergy.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-foxo3-genetic-variants-and-cognitive-performance-in-aged-individuals

Association between FOXO3 genetic variants and cognitive performance in aged individuals

Last updated:
ID:
34991
Start date:
18 May 2018
Project status:
Closed
Principal investigator:
Dr Ashley Webb
Lead institution:
Brown University, United States of America

The goal of the proposed research is to determine if variants in a particular gene, FOXO3, are associated with cognitive function in aged individuals. FOXO3 is a conserved regulator of aging and longevity. In several published studies, single nucleotide polymorphisms in FOXO3 have been linked to longevity and human intelligence. Our study will address the unanswered question of whether these SNPs are linked to cognitive performance in the elderly. This work will provide insight into whether allele status that is associated with longevity is is connected to preservation of cognition with age. This work aligns with the mission of the UK Biobank because it will contribute to our understanding of the genetic factors affecting cognitive function in the elderly. Decline in cognitive function is a hallmark of aging and a major feature of neurodegenerative disease, but the underlying causes remain incompletely understood. Understanding the link between genetic factors that promote healthy aging and preserve learning and memory in aged and diseased individuals has the potential to promote health throughout society. We will extract SNP information for each participant at the FOXO3 locus and determine SNP frequency. We will then bin the population by age and determine SNP frequencies in each group. We will then correlate these SNP frequencies with intelligence response scores from 13 intelligence questions answered by each participant. We will determine if there is a decline in performance on these tests with age independent and/or correlated with FOXO status. The full cohorts are needed. The complete dataset is necessary to evaluate the association between FOXO3 SNPs and cognitive performance with age.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-frailty-risk-and-biological-age

Association between frailty risk and biological age

Last updated:
ID:
653978
Start date:
15 July 2025
Project status:
Current
Principal investigator:
Professor Li Xiangwei
Lead institution:
Shanghai Jiao Tong University School of Medicine., China

1. Research Questions! What is the relationship between biological age and frailty risk in aging populations? How do different types of biological age metrics (e.g., epigenetic age, metabolic age, and phenotypic age) predict frailty risk compared to chronological age? Can biological age serve as a reliable early marker for identifying individuals at higher risk of developing frailty?
2. Research Objectives! To investigate the association between biological age and frailty risk, identifying which biological age metrics most accurately predict frailty;
This study will contribute to understanding the role of biological aging in frailty, offering potential tools for early screening and personalized interventions. The findings could advance health monitoring and prevention strategies, supporting healthier aging and reducing the burden of frailty on healthcare systems.
3. scientific rationale
Frailty is a critical geriatric syndrome characterized by increased vulnerability to adverse health outcomes, including disability, hospitalization, and mortality. Traditional risk assessments often rely on chronological age, which does not fully capture individual variations in aging and frailty progression. Biological age, a composite measure reflecting physiological and molecular changes associated with aging, has emerged as a more precise indicator of health status and disease risk. Multiple biological aging metrics, including epigenetic age, metabolic age, and phenotypic age, have been proposed as potential predictors of frailty. However, their relative utility in assessing frailty risk remains unclear. By leveraging large-scale data from the UK Biobank, this study aims to systematically evaluate the relationship between biological age and frailty risk, identifying the most effective biological aging metrics for predicting frailty onset and progression.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-genetic-lifestyle-environmental-and-clinical-determinants-and-noncommunicable-diseases

Association between Genetic, lifestyle, environmental, and clinical determinants and noncommunicable diseases

Last updated:
ID:
191644
Start date:
24 October 2024
Project status:
Current
Principal investigator:
Dr Junkang Zhao
Lead institution:
Shanxi Bethune Hospital, China

Noncommunicable diseases (NCDs), also known as chronic diseases, tend to be of long duration and are the result of a combination of genetic, physiological, environmental, and behavioral factors.
The main types of NCDs are cardiovascular diseases (such as heart attacks and stroke), cancers, chronic respiratory diseases (such as chronic obstructive pulmonary disease and asthma), autoimmune diseases, and metabolic diseases. NCDs kill 41 million people each year, equivalent to 74% of all deaths globally. Each year, 17 million people die from an NCDs before age 70.

In previous studies, it is well-known that chronic diseases are the result of a long-term combination of genetic predisposition, environmental factors, physiological conditions, and behavior patterns, which could further affect the prognosis of chronic diseases NCDs. Previous studies have made some efforts on risk factors, early diagnosis, or prognosis of chronic diseases, but results are often different and warrant further investigation in large cohorts such as the UK Biobank!
Therefore, this study aims to integrate comprehensive data, such as genetic, environmental, behavioral factors, laboratory indicators, imaging data, and omics data, to discover potential risk factors contributing to the onset and development of chronic diseases, and fit optimal models to show which factors could improve risk prediction, be potential biomarkers for early diagnosis or survival assessment, and what mechanisms may explain these effects. In addition, we believe genetic variation may explain the relationship between exposed factors and NCDs. Using Mendelian Randomization, we’ll examine if there’s a causal link.

We intend to carry out our research with a three-year duration.
The expected outcome is to fill gaps in existing research through comprehensive studies, to provide new and stronger scientific evidence for chronic NCDs prevention, and health management, and to inform policies and interventions that aim to promote health equity and reduce the burden of NCDs.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-genetic-variants-and-incidence-of-cardiac-arrhythmias-among-patients-with-ischemic-and-nonischemic-cardiomyopathy-in-the-uk-biobank-population

Association between genetic variants and incidence of cardiac arrhythmias among patients with ischemic and nonischemic cardiomyopathy in the UK Biobank population

Last updated:
ID:
68952
Start date:
19 April 2021
Project status:
Closed
Principal investigator:
Dr Jim Cheung
Lead institution:
Weill Cornell Medical College, United States of America

Heart failure today remains one of the leading causes of morbidity and mortality worldwide (Mozafarrian et al. 2016). Indeed, despite significant advances in therapy, heart failure continues to carry a 5 -year mortality rate approaching 50%. As heart failure progresses, buildup of scar tissue in the heart increases the risk of unstable electrical rhythms (arrhythmias) which can be fatal. Although electrophysiological interventions such as defibrillator insertion can help lengthen mortality, it remains challenging to identify which patients are at highest risk and therefore stand to benefit the most from invasive procedures. Furthermore, in many instances the arrhythmias may be driving the heart failure in the first place, and addressing them earlier may offer significant benefit.

Importantly, recent studies have shown that certain genetic mutations may increase the risk of both heart failure and cardiac arrhythmias, though the interplay between them remains unclear. Many of these studies have been limited by relatively small sample size. Fortunately, the UK Biobank is a robust dataset that has longitudinally followed hundreds of thousands of patients which can provide the statistical power to help shed light on these previously unanswered questions.

By investigating the associations between genetic variants, heart failure, and cardiac arrhythmias, we can help identify which patients are at highest risk of developing cardiac arrhythmias and help providers to target interventions accordingly. This study would also help determine whether upfront genetic sequencing of patients may be of value in prognostication and risk stratification, which is currently not the standard of care given that these associations remain unclear. This would ultimately be the first study of its kind, and we hope to meet the goal of the UK Biobank and deliver significant value to public health.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-hearing-aids-and-cognitive-function-among-older-adults-at-increased-risk-for-dementia

Association between hearing aids and cognitive function among older adults at increased risk for dementia

Last updated:
ID:
522631
Start date:
5 November 2024
Project status:
Current
Principal investigator:
Dr Yanan Luo
Lead institution:
Peking University, China

Hearing loss (HL) and dementia are both crucial causes of disability among older adults, which seriously threatened the quality of life and brought about huge disease burden on family and society. Hearing aids rehabilitation is of great significance in reducing the risk of dementia. According to the latest report from the Lancet Commission, eliminating hearing loss will reduce the incidence of dementia by 7%. Studies have indicated that hearing aid users have a lower risk of developing mild cognitive impairment (MCI) compared to non-users. However, existing evidence is scarce on the effect of hearing aids on cognitive function among older adults with increased dementia risk, considering that those at high risk of dementia may face more difficulties, such as mastering knowledge and skills in hearing aid use, passing the adaptation period, and maintaining hearing aid use behavior. Therefore, we aimed to explore the status quo of hearing aid use and clarify the association between hearing aids and cognitive function among older adults with high risk for dementia. To be specific, we aimed to explore the utilization rate of hearing aids among older adults with HL and compare the adherence in hearing aid use between those with and without high risk for dementia. We also aimed to clarify the association between hearing aid use and cognitive function, including global cognition, episodic memory, and executive function. We also aimed to examine the association between hearing aid use and the risk of MCI and dementia incidence, along with the transition from MCI to dementia among older adults with high risk for dementia.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-high-penetrance-cancer-susceptibility-mutations-and-clonal-hematopoiesis

Association between high penetrance cancer susceptibility mutations and clonal hematopoiesis

Last updated:
ID:
72687
Start date:
6 October 2021
Project status:
Current
Principal investigator:
Dr Kelly Leigh Bolton
Lead institution:
Washington University in St. Louis, United States of America

Recent studies in healthy individuals have shown that as we age, it is common to develop changes in our DNA. This commonly occurs in the blood and is called clonal hematopoiesis (CH). CH is associated with an elevated risk of blood cancers, cardiovascular disease and other diseases. We know that the likelihood of developing CH is in part inherited (passed down from our parents) . Understanding how inherited genetics influences CH would provide insight into the biology of the earliest stages of blood cancer formation. Here we will use the UK Biobank data to identify genes that predispose healthy people to develop CH.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-hospital-admissions-and-inflammation-on-cognitive-decline-and-neurodegeneration-in-older-adults

Association between hospital admissions and inflammation on cognitive decline and neurodegeneration in older adults

Last updated:
ID:
82574
Start date:
22 March 2022
Project status:
Current
Principal investigator:
Professor Robert Sanders
Lead institution:
University of Sydney, Australia

With the rapid ageing of the world’s population, research to optimise the cognitive health of older adults is vital to maintaining their independence and social participation. This project aims to improve our understanding of the impact of hospitalisation on changes in cognition otherwise observed in healthy ageing. The objective of this study is to identify the mechanisms and modifiable risk factors, associated with acute illness, to minimize harm and inform future healthcare innovation.
We will analyse data from the UK Biobank to assess changes in cognition in this cohort associated with cumulative hospital admissions, as a measure of cumulative burden of acute illness. We will adjust our analyses for known risk factors like ageing, cardiovascular health and alcohol. However, we also seek to contribute new findings on associations hospital admissions, brain degeneration, injury to white matter tracts in the brain and inflammation to test whether these are potential mechanisms for the changes in cognitive health over time. Hospital admissions will be analyzed using the established data registry of Hospital Episode Statistics. We will utilize data on cognitive health in this cohort using the touchscreen and online follow-up standardized cognitive testing and compare these results to lifestyle factors collected at the same time and over multiple timepoints. Results of existing blood tests and brain imaging will then be used to evaluate plausible associations between hospital admissions and brain injury and inflammation.
This project is anticipate to take 36 months from receiving the data to publication submission and will involve two data scientists and two senior investigators experienced in dementia and medical research.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-ketohexokinase-mutations-and-adverse-cardiovascular-risk-factors-and-outcomes

Association between ketohexokinase mutations and adverse cardiovascular risk factors and outcomes

Last updated:
ID:
43833
Start date:
25 June 2019
Project status:
Current
Principal investigator:
Dr Joseph A Johnston
Lead institution:
Eli Lilly and Company Ltd (USA), United States of America

Essential fructosuria (EF) is an asymptomatic condition caused by those mutations in the ketohexokinase (KHK) gene that lead to loss of the ability to metabolize fructose. EF occurs with a frequency of 1 in 100,000 to 130,000 individuals who have mutations in both copies of the KHK gene, resulting in an abnormal KHK protein. Given the high concentration of fructose in the typical Western diet, blocking the function of KHK and the normal processing of fructose is an intriguing potential target for drug development. Our proposal is to use the genetic data from the UK Biobank to identify a cohort of individuals with (likely undiagnosed) EF and compare the cardiovascular and metabolic profile of these individuals to that of a matched cohort of individuals without these KHK mutations. Cohorts will be compared on the basis of both concurrently measured outcomes, such as body mass index and lipid levels, as well as long-term clinical outcomes, including the onset of diabetes and/or ischemic heart disease. Results suggesting that human KHK “knock-outs” have a favorable cardio-metabolic profile could help validate KHK as a therapeutic target and spur development of novel medicinal agents.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-lifestyle-dietary-patterns-and-neurological-and-spinal-diseases

Association between Lifestyle, Dietary Patterns, and Neurological and Spinal Diseases.

Last updated:
ID:
162345
Start date:
5 January 2024
Project status:
Current
Principal investigator:
Dr Shaobin Feng
Lead institution:
Shandong Provincial Hospital, China

Neurological and spinal disorders are highly prevalent in our society, casting a substantial economic burden on both communities and families. Primary and secondary prevention strategies are of paramount importance. To date, there has been substantial research exploring the potential connection between diet, lifestyle, and the health of the nervous system and spine. Our objective is to conduct this study to shed light on the role of lifestyle and dietary habits in promoting neurological and spinal health.

The UK Biobank is a comprehensive, large-scale cohort study that enrolled about 500,000 participants from 2006 to 2010. It meticulously gathered extensive data on participants’ lifestyles and dietary practices while continuously monitoring the occurrence of neurological and spinal disorders. Previous studies have documented correlations between common lifestyle factors like physical activity, diet, smoking, and alcohol consumption and the risk of various neurological and spinal disorders. However, contemporary society is witnessing the emergence of new lifestyle trends and dietary preferences, such as the increasing popularity of shift work and vegetarianism.

Prior research has indicated that shift work may exert detrimental effects on cardiovascular health, skeletal well-being, and liver conditions, while vegetarians may be at a heightened risk of anemia and fractures. Consequently, further research into the impact of these factors on neurological and spinal disorders holds significant importance.

The entire project is planned to span approximately three years. Through a comprehensive analysis of the extensive dataset available from the UK Biobank, we have the opportunity to pinpoint potential risk factors and intervention strategies, ultimately enhancing people’s quality of life and reducing the incidence of neurological and spinal diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-liver-diseases-and-neurodegenerative-diseases

Association between liver diseases and neurodegenerative diseases

Last updated:
ID:
604141
Start date:
17 April 2025
Project status:
Current
Principal investigator:
Dr Wu Yu
Lead institution:
Yale University, United States of America

Lay Summary

Liver diseases, such as Metabolic dysfunction-associated steatotic liver disease (MASLD), drug-induced liver injury (DILI), and hepatocellular carcinoma (HCC), among others, pose significant public health challenges due to their systemic effects beyond the liver. Increasing evidence suggests that liver dysfunction may play a role in the development of neurodegenerative diseases, such as Alzheimer’s disease, Parkinson’s disease, and vascular dementia. These associations may be driven by shared metabolic, inflammatory, and genetic mechanisms.

This study aims to explore the relationship between a broad spectrum of liver diseases and the risk of neurodegenerative conditions, leveraging the extensive data available in the UK Biobank. Specifically, we will investigate whether liver disease increases susceptibility to cognitive decline and neurodegeneration, identify potential genetic and metabolic mediators, and assess risk variability across different types and severities of liver diseases.

Understanding these connections will advance our knowledge of the systemic consequences of liver diseases and inform strategies for early intervention and integrated care. Additionally, this research will quantify the burden of neurodegenerative diseases linked to liver disease, contributing to the development of comprehensive healthcare strategies that address both liver and brain health.

Scientific Questions

Are liver diseases (e.g., MASLD, DILI, HCC) associated with an increased risk of neurodegenerative diseases such as Alzheimer’s disease, Parkinson’s disease, and vascular dementia?
What roles do genetic, metabolic, and inflammatory factors play in mediating the relationship between liver diseases and neurodegenerative diseases?
Are specific subtypes or severities of liver diseases associated with a greater risk of neurodegenerative diseases?
How does the duration or progression of liver disease influence the risk and severity of neurodegeneration?


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-long-term-medication-use-and-ageing

Association between long-term medication use and ageing

Last updated:
ID:
90471
Start date:
20 December 2022
Project status:
Current
Principal investigator:
Dr Erin Kelty
Lead institution:
University of Western Australia, Australia

Long-term medication use may affect how we age. We will examine how commonly used medicines, such as those used to treat chronic pain, cardiovascular disease, and diabetes, are associated with biological indicators of age. Biological indicators of age such as laboratory tests, genetic markers, imaging, and physical measures can be used to tell if a person’s body is older or younger than their actual or chronological age.
For this study we are utilising UK Biobank data to examine participants who have used specific medicines and how this relates to biological indicators of aging compare to their chronological age. We will also compare how different medicines used to treat the same condition may differ in terms of their association with biological indicators of aging. Having a good understanding of the long-term impacts of different medicines on ageing is important to ensure the health of people treated with these medicines, especially as many countries continue to see an increasing aging population.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-metabolic-dysfunction-associated-steatotic-liver-disease-masld-and-extrahepatic-diseases

Association between metabolic dysfunction-associated steatotic liver disease (MASLD) and extrahepatic diseases

Last updated:
ID:
170239
Start date:
20 March 2024
Project status:
Current
Principal investigator:
Dr Tengyan Wu
Lead institution:
Guangxi Medical University, China

Lay summary
Metabolic dysfunction-associated steatotic liver disease (MASLD) has established a new diagnostic standard for fatty liver disease independent of alcohol intake and associated viral hepatitis infection. MASLD is different from the previous nonalcoholic fatty liver disease (NAFLD) and is defined by its own set of inclusion criteria rather than exclusion criteria. NAFLD has a certain correlation with extrahepatic diseases. However, it is not clear whether the newly defined MASLD is related to extrahepatic diseases. Therefore, we plan to explore the relationship between MASLD and extrahepatic diseases using the data of the British Biological Bank, and build a prediction model of related diseases. In addition, we will use the cohort study data of a single center to verify (Registration number!ChiCTR2200058543). This study will be of great significance to the early intervention and treatment of MASLD. More importantly, we are more interested in exploring the burden of disease caused by MAFSLD.

Scope extension: Scientific Questions:

1) Is MASLD associated with long-term extrahepatic and extrahepatic cancer?
2) Is MASLD associated with long-term cardiovascular and cerebrovascular diseases?
3) Is MASLD associated with long-term chronic kidney disease?
4) Is MASLD associated with other long-term systemic diseases?
5) How much of a disease burden does MASLD impose?

Aim:

1) To explore the long-term prognosis of MASLD patients, including cancer incidence rate, cardiovascular events and mortality.
2) To explore the relationship between the incidence rate of different systemic diseases in patients with MASLD with different gene phenotypes.
3) To explore the factors affecting the occurrence of intrahepatic and extrahepatic diseases in patients with MASLD.
4) To build a prediction model for the occurrence of intrahepatic and extrahepatic diseases in patients with MASLD.
5) To measure the burden of disease caused by MASLD.

Time: 2024/01/01-2026/12/31


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-modifiable-lifestyle-factors-and-onset-of-multiple-long-term-conditions-with-a-view-to-causality

Association between modifiable lifestyle factors and onset of multiple long term conditions, with a view to causality

Last updated:
ID:
529562
Start date:
4 March 2025
Project status:
Current
Principal investigator:
Dr Alexander Pate
Lead institution:
University of Manchester, Great Britain

The development of multiple long-term conditions (MLTCs, previously referred to as multimorbidity) is a priority for global health research. The complex relationship between lifestyle factors and MLTCs remains largely unexplored. Drug based interventions (e.g. statins) often focus on the risk reduction for single outcomes (e.g. prevention of cardiovascular disease). In contrary, many lifestyle factors (e.g. exercise) will have a preventative effect on more than one outcome (e.g. cancer, cardiovascular disease and diabetes). Unpicking the relationship between lifestyle factors and MLTCs is therefore of interest to take a more holistic view of patient care, in a time where individuals living with multiple long term-conditions is rising. The aim of this project is to explore the relationship between lifestyle factors and occurrence of multiple long-term conditions (MLTC) in the UK population.

Objective 1: Clustering Analysis to identify key relationships between lifestyle factors and multiple long term health conditions (MLTCs)

Apply clustering techniques: Apply clustering techniques to group the MLTCs and validate. Supervised algorithms will be trained on lifestyle factors (physical activity, sleep, smoking, diet, alcohol consumption and sun exposure).

Objective 2: Association Analysis for key relationships
Univariable and multivariable analyses, estimating association between lifestyle factor and multimorbid groups (and contributing conditions), quantified through risk difference, risk ratio and odds ratios.

Objective 3: Causal Analysis for key relationships
Directed Acyclic Graphs (DAGs): DAGs will be constructed to help identify confounding variables and motivate adjustment sets. We will use causal modelling techniques (standardisation, inverse probability weighting) to estimate the average treatment effects. Target trial emulation frameworks will be used where relevant. Direct effects will be estimated using mediation analysis.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-multi-level-biomarkers-lifestyles-environmental-factors-and-their-interaction-in-the-occurrence-and-progression-of-chronic-non-communicable-diseases

Association between multi-level biomarkers, lifestyles, environmental factors and their interaction in the occurrence and progression of chronic non-communicable diseases

Last updated:
ID:
310446
Start date:
10 April 2025
Project status:
Current
Principal investigator:
Dr Yuancheng Li
Lead institution:
Institute of Dermatology, CAMS, China

All efforts are expected to be finished in the next 36 months. Our research endeavors to present valuable evidence that will propel the advancement of personalized prevention measures for NCDs, thereby enhancing the overall effectiveness of healthcare services tailored to individual needs.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-multi-omic-features-and-various-phenotypes-for-identification-of-biomarkers-and-therapeutic-targets-for-complex-brain-disorders

Association between multi-omic features and various phenotypes for identification of biomarkers and therapeutic targets for complex brain disorders.

Last updated:
ID:
314730
Start date:
9 January 2025
Project status:
Current
Principal investigator:
Professor Yongyong Shi
Lead institution:
Shanghai Jiao Tong University, China

Based on the genotype and phenotype data of complex brain disorders in the UK Biobank, we aim to explore the etiological mechanisms involving genetic-environmental interactions, and identify potential therapeutic targets for complex brain disorders.
Utilizing causal inference methods, we can delve into associations between complex brain disorders or associations between diseases and non-disease phenotypes, aiming to unravel the mechanisms behind the occurrence of complex diseases/traits. We will integrate multi-level and multimodal genetic data, including WGS, WES, imaging data, and metabolomic data, to explore the proportional impact of genetic and environmental factors on the onset of complex diseases/traits. Data obtained from the UK Biobank would be combined with data from other cohorts or data of clinical samples attained from third-generation sequencing / single-cell sequencing / spatial omics sequencing / etc., which can contribute to more reliable research conclusions.
The estimated duration of this project is 36 months.
This project is anticipated to provide more targeted strategies for future treatments and interventions, as well as new biomarkers for early intervention and precise treatment, for complex brain disorders.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-neighbourhood-walkability-and-physical-activity-behaviours-in-europe

Association between neighbourhood walkability and physical activity behaviours in Europe

Last updated:
ID:
714369
Start date:
26 June 2025
Project status:
Current
Principal investigator:
Mrs Thi Hoang Ha Nguyen
Lead institution:
Amsterdam UMC Research BV, Netherlands

The benefits of physical activity (PA) for health is well established, and include reductions in cardiometabolic risk factors and mortality rates, as well as improvements in mental health and quality of life (1-4). However, more than a quarter of adult population fails to meet the World Health Organisation’s PA guidelines (5).

European countries exhibit relatively high levels of physical inactivity, though a wide variety exists both between and within countries. Guthold et al. suggest that PA level is influenced by national, subnational, and community-level factors (5), indicating the need to explore contextual determinants of PA. Among these, built environment characteristics – such as neighbourhood walkability – play a crucial role in promoting PA. Numerous studies have reported positive associations between higher walkability in urban areas and increased PA, whether through active transportation (e.g., walking, cycling) or other forms of exercise during work and leisure time (6-10).

Walkability incorporates multiple environmental components, such as street connectivity, land use mix, residential density, public transport density, and number of parks (11, 12). Combination of multiple variables explains the impact of neighbourhood characteristics better than single-variable measures alone (12).

Patel et al. recently developed a comprehensive walkability index to a European scale, at high resolution (100 × 100 m grids), with geospatial innovations to capture multiple environmental factors. This index included seven components, including street walk length, number of street intersections, green spaces, slope, public transport stops, land use mix, and area of isochrones.

This study will utilise this walkability index to examine the association between neighbourhood walkability and physical activity behaviours among adults across various European regions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-neurotropic-herpes-viruses-and-early-signs-of-alzheimers-disease-impact-of-susceptibility-factors

Association between neurotropic Herpes viruses and early signs of Alzheimer’s disease: impact of susceptibility factors

Last updated:
ID:
99119
Start date:
4 May 2023
Project status:
Current
Principal investigator:
Dr Catherine Helmer
Lead institution:
University of Bordeaux, France

The aim of this project is to investigate the role of infections, particularly those due to Herpes viruses, in Alzheimer’s disease.
Alzheimer’s disease is a devastating condition with consequences for affected people, their family and the society. Although several risk factors of Alzheimer’s disease have been identified, to date its causes remain misunderstood. In the past years, results from several researches have suggested that infections, especially those related to Herpes viruses, may be involved in the occurrence of Alzheimer’s disease. Herpes viruses have the ability to stay in the body in a latent stage after a first infection, and to reactivate periodically. Several studies suggested that these viruses could promote the development of Alzheimer’s disease lesions in the brain.
This project will leverage the wealth of the UK Biobank data where a biological measure of infectious history against several viruses, including Herpes viruses, has been performed in about 10 000 participants. We will analyze whether a history of infection against Herpes viruses (taken separately or combined) or a high number of prior infections (considering the biological measure of all the infections) are associated with a greater risk of cognitive decline (like memory decline) or changes in the brain (thanks to Magnetic Resonance Imaging data). In addition, to fully understand which persons are most at risk of developing Alzheimer’s disease, we will study potential susceptibility factors, i.e. factors that could modulate the impact of Herpes viruses on the brain (such as age, genetics, factors impacting immune defenses and co-infections). Finally, among infected participants we will assess whether those taking anti-viral medications have a lower risk of cognitive decline and brain changes.
We anticipate 36 months from the reception of UK Biobank data to project completion.
This project will improve the understanding of Alzheimer’s disease, by clarifying the potential impact of infectious diseases. This could open new perspectives of preventive strategies to delay or avoid the occurrence of Alzheimer’s disease (including vaccines, anti-viral treatments!) and help to better target participants to be included in future trials (targeting the most at-risk participants who could beneficiate from treatments).


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-night-shift-work-and-nafld

Association between night shift work and NAFLD

Last updated:
ID:
79302
Start date:
8 December 2021
Project status:
Current
Principal investigator:
Dr Hangkai Huang
Lead institution:
Zhejiang University, China

Non-alcoholic fatty liver disease (NAFLD) has emerged as the most common chronic liver disease, affecting nearly one third of adults worldwide. The climbing prevalence of NAFLD was fueled by the rapid rise in obesity, type 2 diabetes, cardiovascular disease and other cardiometabolic diseases. NAFLD patients are associated with increased risks of hepatocellular carcinoma as well as extra-hepatic cancers. The burden of NAFLD prevalence has posed a big challenge to global health resources. It is urgent to explore the way of timely prevention and intervention of NAFLD. Mounting evidence has confirmed that shift work posed negative impact on individual health. In particular, long-term night shift work was associated with the increased risks of diabetes, atrial fibrillation, ischemic stroke and other cardiometabolic diseases. However, the association between night shift work and NAFLD was unclear. This proposed study aims to explore whether individuals with night shift work carry higher risks of developing NAFLD. This study will potentially provide evidence of the adverse impact of night shift work on NAFLD and reveal new approaches to prevent the development and progression of NAFLD.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-night-shift-work-mental-health-status-and-coronary-heart-disease-risk-a-prospective-cohort-study-using-uk-biobank-data

Association Between Night Shift Work, Mental Health Status, and Coronary Heart Disease Risk: A Prospective Cohort Study Using UK Biobank Data

Last updated:
ID:
910056
Start date:
27 September 2025
Project status:
Current
Principal investigator:
Professor Yongle Li
Lead institution:
General Hospital, Tianjin Medical University, China

Research Questions
1. Does long-term night shift work independently increase coronary heart disease (CHD) risk?
2. Do mental health disorders mediate the association between night shift work and CHD?
3. Are there genetic predispositions that modify this relationship?
Objectives
1. Primary: Quantify the association between cumulative night shift work exposure and CHD incidence using UK Biobank data.
2. Secondary:
(1) Assess mediation effects of depression/anxiety on night shift-CHD pathway
(2) Identify vulnerable subgroups through effect modification analysis
(3) Establish causal relationships via Mendelian randomization using genetic instruments
Scientific Rationale
Night shift work affects 20% of the workforce and disrupts circadian rhythms, potentially increasing cardiovascular risk through multiple pathways: altered autonomic function, inflammatory responses, and metabolic dysregulation. Mental health disorders, prevalent among shift workers, independently predict CHD and may represent a critical mediating mechanism.
Current evidence remains inconclusive due to methodological limitations and insufficient adjustment for confounders. This study addresses these gaps through prospective design, comprehensive covariate adjustment, and genetic approaches to strengthen causal inference.
Expected impact includes informing occupational health policies, developing risk stratification tools for shift workers, and identifying intervention targets to reduce CHD burden in this vulnerable population.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-non-invasive-markers-of-liver-fibrosis-and-chronic-kidney-disease-in-people-with-type-2-diabetes-and-the-role-of-pnpla3-tm6sf2-gene-variants-cross-sectional-analysis

Association between non-invasive markers of liver fibrosis and chronic kidney disease in people with type 2 diabetes and the role of PNPLA3, TM6SF2 gene variants: cross-sectional analysis

Last updated:
ID:
56800
Start date:
31 March 2020
Project status:
Current
Principal investigator:
Professor Chris Byrne
Lead institution:
University of Southampton, Great Britain

This study aims to look at the link between non-alcoholic fatty liver disease (NAFLD) and chronic kidney disease in people with type 2 diabetes. NAFLD is thought to affect as much as 25% of the global population mainly because of the growing international obesity problem. NAFLD can lead to severe scarring of the liver called ‘cirrhosis’. In some cases people with NAFLD can even develop liver cancer. People with diabetes are at the highest risk of getting NAFLD. People with diabetes also seem to be at the highest risk of getting scarring due to NAFLD. There also seem to be some genetic factors which put people at more risk of getting liver scarring due to NAFLD.

However, NAFLD does not just affect the liver in people with diabetes. NAFLD affects other organs as well. For example, NAFLD seems to have a strong link with chronic kidney disease. People with diabetes are known to be at risk of chronic kidney disease. However, we think that people with diabetes who have NAFLD might be at even more risk of chronic kidney disease. Current guidelines do not recommend considering NAFLD as an extra risk factor when treating patients with diabetes who have chronic kidney disease. In this study we aim to confirm the link between NAFLD and chronic kidney disease in people with diabetes using the large data resource of patient information called UK Biobank. If a link between NAFLD and chronic kidney disease is confirmed then it might have implications for the treatment of people with diabetes who have either NAFLD or chronic kidney disease. The presence of a link between the two diseases would suggest that when we are treating a patient with NAFLD it is also important to consider their kidneys and the risk of chronic kidney disease. By knowing this risk exists then it is possible that treatment could be started to prevent complications before they happen. We aim to complete this project over 24 months with publication of our findings at the end of that timeframe.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-obesity-and-osteoarthritis-plasma-proteomics-and-causal-inference-in-the-uk-biobank

Association between obesity and osteoarthritis: plasma proteomics and causal inference in the UK Biobank

Last updated:
ID:
905784
Start date:
24 July 2025
Project status:
Current
Principal investigator:
Dr Yudan Wang
Lead institution:
North China University of Science and Technology, China

Scientific rationale!Obesity is a key modifiable osteoarthritis (OA) risk factor involving both biomechanical and metabolic-inflammatory mechanisms, though exact molecular pathways remain unclear. Large-scale plasma proteomic profiling offers a powerful approach to characterize systemic metabolic dysregulation, inflammatory signatures, and tissue remodeling processes associated with obesity-related OA pathogenesis. The UK Biobank’s extensive proteomic data, when combined with its rich phenotypic and genomic resources, provides a unique opportunity to identify novel protein biomarkers and elucidate disease mechanisms. Furthermore, established genetic associations from genome-wide association studies (GWAS) enable the application of Mendelian randomization (MR) approaches to strengthen causal inference while minimizing confounding, particularly through the use of obesity-associated genetic variants as instrumental variables.
Aims!(1) Quantify the independent contributions of mechanical loading (evaluated by BMI and body composition) versus metabolic-inflammatory pathways (assessed through adipokines, systemic inflammatory markers, and fat distribution patterns) to OA pathogenesis; (2) Characterize effect modification by sex, age, and other demographic/clinical factors in the obesity-OA relationship using stratified and interaction analyses; (3) Systematically identify obesity-associated plasma protein signatures and their implicated biological pathways implicated in OA development through large-scale proteomic profiling; (4) Employ GWAS and MR frameworks to both determine the causal effect of obesity on OA risk and investigate proteomic mediators of this relationship through mediation MR-based mediation analyses. Public health impact!This study will predict OA risk in obesity, evaluate weight management benefits, and uncover obesity-OA mechanisms to guide targeted prevention and reduce disease burden.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-ocular-vascular-diseases-and-systemic-conditions

Association between ocular vascular diseases and systemic conditions

Last updated:
ID:
649353
Start date:
1 April 2025
Project status:
Current
Principal investigator:
Dr Jinfeng Cao
Lead institution:
Second Hospital of Jilin University, China

Background: Ocular vascular diseases, such as diabetic retinopathy, age-related macular degeneration, and retinal vein occlusion, are linked to systemic vascular dysfunction and lifestyle factors. These diseases often co-occur with conditions like diabetes, hypertension, and sleep disturbances. Investigating these associations can reveal shared vascular and inflammatory mechanisms, improving early detection and management strategies for at-risk individuals.

Objective: To study the associations between ocular vascular diseases and systemic conditions (e.g., cardiovascular health, inflammation) and lifestyle factors (e.g., sleep quality, physical activity), and to identify potential biomarkers or modifiable risk factors.

Methods: This study will use the following UK Biobank data for a cross-sectional and longitudinal analysis:
1. Ocular data: OCT and OCT-A for macular and choroidal thickness, vascular density, and etc.
2. Systemic data: Carotid ultrasound (intima-media thickness, flow), HbA1c, CRP, lipid profiles, blood pressure and etc.
3. Lifestyle and sociodemographic data: Sleep questionnaires, actigraphy, physical activity levels, smoking, alcohol consumption, and etc.
4. Genetic data: Genome-wide genotyping data for hundreds of thousands of single nucleotide polymorphisms (SNPs).
Statistical methods will include regression analyses, GWAS, mendelian randomizationmediation and time-to-event analysis.

Expected outcomes: The research aims to clarify the systemic and lifestyle determinants of ocular vascular diseases, identify shared pathophysiological mechanisms, and suggest potential targets for early intervention or prevention.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-periodontal-diseases-and-hyperuricemia-gout

Association between periodontal diseases and hyperuricemia/gout

Last updated:
ID:
206893
Start date:
12 March 2025
Project status:
Current
Principal investigator:
Dr Ting Yu
Lead institution:
Guangzhou Medical University, China

Both hyperuricemia and periodontal diseases are highly prevalent non-communicable chronic diseases. They share many significant comorbidities such as diabetes, cardiovascular diseases, and chronic kidney diseases. Some scientific evidence has indicated a possible association between hyperuricemia and periodontal diseases. However, the strength of evidence is very weak, and the direction of association is unclear. It is necessary to answer whether there is a higher risk of periodontitis in populations with hyperuricemia/gout or vice versa. Hence, we would like to conduct clinical research based on the UK population using the UK Biobank data. The research might include a cross-sectional design and cohort design. We are prepared to consider three years to conduct the research project and publish the results. If hyperuricemia/gout were found to be positively connected with periodontal diseases or vice versa, given the high prevalence of both conditions and growing incidence of hyperuricemia, the global burden associated with them would be sharply increased. It would also increase the burden of many comorbidities that they share. Then it would shape clinical practice including the treatment regimen of both conditions and their comorbidities.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-phenotypes-of-pharmacogenes-and-drug-related-outcomes

Association between phenotypes of pharmacogenes and drug-related outcomes

Last updated:
ID:
173505
Start date:
27 November 2024
Project status:
Current
Principal investigator:
Dr Cornelia Schneider
Lead institution:
University Hospital Basel, Switzerland

We aim to study the association between a group of genes existing in different variants, that are believed to have an impact on how drugs act in our body and the respective health outcomes. In severe cases, persons with a specific gene variant are at risk of being hospitalized for adverse drug reactions when taking certain drugs – if the treatment is not adapted to their genetic profile. We aim to quantify these effects for different associations between genes, drugs, and health outcomes to provide more precise evidence for clinical practice as to who should be tested for which genes before starting drug therapy. We also aim to provide more evidence on which adverse drug reactions can be expected with certain gene variants. We hope that our research will thus help to reduce adverse drug reactions in the population and improve drug effectiveness. The project is planned for a duration of 3 years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-pregnancy-and-long-term-changes-in-maternal-cardiovascular-imaging-phenotypes-based-on-multi-omics-analysis-with-uk-biobank-data

Association between Pregnancy and Long-term Changes in Maternal Cardiovascular Imaging Phenotypes: based on Multi-Omics Analysis with UK Biobank Data

Last updated:
ID:
615699
Start date:
2 April 2025
Project status:
Current
Principal investigator:
Mr Hao Ying
Lead institution:
Tongji University School of Medicine, China

Research Questions: Pregnancy is a significant physiological event in women’s life course, though its influence on maternal long-term cardiovascular health is still not entirely clear. The aim of this study is to explore how pregnancy-related conditions (i.e. childbearing history & adverse pregnancy outcomes, CBH & APO) affect maternal long-term cardiovascular imaging phenotypes (heart MRI & carotid ultrasound), and to further analyze the roles of multi-omics factors such as genomics, proteomics, metabolomics and exposomics in this case.
Objectives: 1. Reveal the effect of CBH (e.g. whether to give birth, parity, delivery method) and APO (e.g. miscarriage, preterm birth) on maternal long-term cardiovascular imaging phenotypes and their potential causal relationships. 2. Identify omics profiles that increase women’s susceptibility to CVD and investigate mediating roles of multi-omics factors in the above pathway. 3. Verify the strength and specificity of the correlation between the selected cardiovascular imaging phenotypes and various CVD. 4. Establish a CVD risk prediction model specifically for parous women, incorporating pregnancy-related conditions, cardiovascular imaging phenotypes, genetic and omics data to identify female individuals at higher risk of CVD.
Scientific Rationale: During pregnancy, a woman’s body undergoes complicated physiological changes that can have a profound impact on long-term cardiovascular health. Previous studies have shown that APOs are associated with increasing risk of women’s long-term CVD, but specific mechanism remains unclear. In recent years, the rapid development of imaging techniques and multi-omics research methods has provided new opportunities for in-depth exploration of this issue. It is expected to unravel the complex relationship between pregnancy-related conditions and maternal long-term cardiovascular imaging phenotypes, and provide novel perspectives and strategies for the assessment and prevention of women’s CVD.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-serum-fabp4-levels-and-obesity-associated-cancer-incidence

Association between serum FABP4 levels and obesity associated cancer incidence

Last updated:
ID:
248403
Start date:
22 October 2024
Project status:
Current
Principal investigator:
Dr Christopher Strouse
Lead institution:
University of Iowa, United States of America

Obesity has been identified as a risk factor for several types of cancer. However, how obesity causes these cancers is still unknown. If we can identify the ways that obesity can cause certain cancers, we may be able to use this knowledge to design new ways to prevent or treat cancers.

Fat cells in humans release a protein called Fatty Acid Binding Protein type 4 (FABP4) into the bloodstream. Patients with more fat cells (e.g. higher levels of obesity) have higher levels of the protein in their blood. Higher levels of FABP4 have been found to correlate with many of the chronic health problems associated with obesity, like heart disease and diabetes. Some studies have also found that FABP4 levels also correlate with risk of cancers. In this study, we will find how much a patient’s FABP4 level relates to their risk of developing a wide range of obesity associated cancers.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-serum-sex-steroid-hormones-and-ductal-carcinoma-in-situ-among-uk-women

Association between serum sex steroid hormones and ductal carcinoma in situ among UK women

Last updated:
ID:
30247
Start date:
1 August 2017
Project status:
Closed
Principal investigator:
Dr Thomas Rohan
Lead institution:
Albert Einstein College of Medicine, United States of America

Endogenous sex steroid hormone levels, particularly oestrogen, are strongly associated with increased risk of invasive ductal carcinoma, but their role in the development of ductal carcinoma in situ DCIS, which is the earliest form of breast cancer, is unclear. This study, therefore, aims to i) investigate the association of oestrogen, testosterone and SHBG with risk of DCIS among women; ii) assess how these associations are modified by age, menopausal status, and weight. This study aims to improve our understanding of aetiology of early stage ductal carcinoma (DCIS). Such information can be useful in developing approaches for the prevention of breast cancer, and is in keeping with the UK Biobank?s aim to improve prevention of cancer. This study will be conducted by researchers at Albert Einstein College of Medicine, Department of Epidemiology and Population Health. We will use the data on serum oestrogen, testosterone and SHBG measurements for all women within the UK Biobank prospective cohort who developed DCIS and those without any previous history of cancer. We will also extract information on potential confounders such as age, BMI, parity and menopausal status, from the data base. We will analyse the data to investigate the association of sex steroid hormones with DCIS. This study will include all women with oestradiol, testosterone and SHBG measurements but will exclude those with a past history of DCIS or invasive cancer, and also those who report use of hormone replacement therapy at the time of the sex steroid hormone measurements.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-serum-vitamin-d-deficiency-and-the-risk-of-herpes-zoster-a-longitudinal-uk-biobank-study

Association between serum vitamin D deficiency and the risk of herpes zoster: a longitudinal UK Biobank study

Last updated:
ID:
51265
Start date:
13 September 2019
Project status:
Closed
Principal investigator:
Dr Liang-yu Lin
Lead institution:
London School of Hygiene and Tropical Medicine, Great Britain

When our immune system does not work well, we are more vulnerable to getting infections, such as chickenpox and shingles. This virus that causes chickenpox causes lifelong infections, and it cannot be removed. When the virus that causes chickenpox reactivates in adults shingles develops. A common symptom of shingles is a painful skin rash. Some shingles patients may suffer from long-term nerve pain, which will significantly decrease their quality of life. The treatment for pain symptoms is not very effective, and it increases health spending. Therefore, it is important to study what cause shingles, and to find new ways to prevent it.
Vitamin D is produced by the skin after sun exposure, and it is regarded to be an essential element to bone health. Public Health England advises taking vitamin D supplements every day. Recent studies suggest vitamin D has some effect on immunity, and it might help to prevent viral infections. However, we do not know whether vitamin D levels affect the chance of getting shingles. Furthermore, vitamin D levels are not routinely measured and recorded in patients’ GP records. It is very difficult to study vitamin D by using GP records, unless we can find another way of findings low vitamin D levels. We aim to: 1. To describe how many people in the UK are deficient in vitamin D; 2. To investigate whether vitamin D deficiency increases the risk of shingles; 3. To find ways of identifying vitamin D deficiency in GP records using UK Biobank data linked to GP records.
Understanding the proportion of people with vitamin D deficiency in the UK population will help the public health department to develop guidance about vitamin supplementation. If we find that vitamin D deficiencies increase the risk of shingles, this will inform future research into shingles prevention. Furthermore, our work will also help other researchers to use electronic medical records to study vitamin D.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-shift-work-and-cancer-vascular-disease-and-other-diseases

Association between shift work and cancer, vascular disease and other diseases

Last updated:
ID:
431
Start date:
30 June 2013
Project status:
Closed
Principal investigator:
Dr Ruth Travis
Lead institution:
University of Oxford, Great Britain

Shift work and disease in UK Biobank
Shift work, and in particular night work, has been linked in several studies to an increased risk of several common diseases including certain cancers, cardiovascular disease and type 2 diabetes. However, other studies have not found such associations and it is unclear why risk of these diseases might be higher among shift workers. Possible reasons include the harmful effects of disturbed patterns of certain hormones due to electric light at night, shift workers having disturbed sleep or shift workers being more likely to have known lifestyle risk factors for disease. To better understand the possible relationship between shift work and disease, we aim to compare the characteristics of UK Biobank participants who have and have not done shift work or night work. In subsequent phases of the project we propose to examine the relationships between shift work, sleep and subsequent risks of breast cancer, prostate cancer, cardiovascular disease, diabetes and death. These phases will be conducted when at least 2000 incident cases of each endpoint have accrued in UK Biobank. This project will contribute towards a fuller understanding of a potentially important and modifiable occupational risk factor for several common diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-shift-work-sleep-disorder-and-risks-of-cancers-and-chronic-kidney-disease

Association between shift work, sleep disorder, and risks of cancers and chronic kidney disease

Last updated:
ID:
64314
Start date:
28 October 2020
Project status:
Closed
Principal investigator:
Dr Mark Purdue
Lead institution:
National Cancer Institute, United States of America

Night shift work has been classified as a probable carcinogen (cancer-causing substance) to humans in a recent expert review of the scientific literature convened by the International Agency for Research on Cancer (IARC) Monographs On the Identification of Carcinogenic Hazards to Humans (1). There are inconsistent findings from epidemiologic studies whether shift work plays a key role in increasing risk of various cancers such as cancers of breast, prostate, and colon/rectum (2-8). Animal studies also have shown that constant exposure to light increased incidences of liver cancer, lung cancer, and skin cancer (9, 10). IARC has not yet evaluated the evidence regarding the relationship between sleep disorders and cancer risk, however, it has been shown that sleep apnea and sleep-disordered breathing are important factors for cancer risk and cancer death (12-16). There is limited but increasing evidence that both shift work and sleep disorders reduce kidney function and increase risk of chronic kidney disease (17-21). Patients having sleep-related breathing problems usually do not have enough oxygen in their tissues and may develop chronic kidney disease (22). Thus, we propose to conduct epidemiologic studies to investigate whether shift work or sleep disorder is associated with an increased risk of selected cancers or chronic kidney disease. Findings from our proposed study will provide new evidence regarding the relationships between shift work, sleep disorders and risks of several cancers, as well as chronic kidney disease, which will inform upcoming IARC meeting to evaluate whether they are important risk factors of developing cancers.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-sleep-patterns-of-morningness-eveningness-and-changes-in-cognitive-function

Association between sleep patterns of morningness-eveningness and changes in cognitive function

Last updated:
ID:
94034
Start date:
16 February 2023
Project status:
Current
Principal investigator:
Professor Daqing Ma
Lead institution:
Imperial College London, Great Britain

The aim of the research project is to investigate the association between sleep patterns with cognition.
As current studies have shown an association between sleep duration and quality with cognition, we would like to look if, under the same duration and quality, the pattern of one’s sleep would have an effect on his/her cognitive function.
The project will take around 6 months.
For public health, the findings of this study would yield a meaningful outcome as there are groups of people who specifically work as an eveningness schedule. Hence, the resulted association would be significant to both researcher, public, employers, and employees.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-social-determinants-of-health-and-chronic-non-communicable-diseases

Association between social determinants of health and chronic non-communicable diseases

Last updated:
ID:
116198
Start date:
22 September 2023
Project status:
Current
Principal investigator:
Professor Bin Luo
Lead institution:
LanZhou University, China

Chronic non-communicable diseases (CNCDs), including cardiovascular diseases, cancers, chronic respiratory diseases, diabetes, and mental disorders are a major public health challenge, accounting for 71% of global deaths and 85% of premature mortality in low- and middle-income countries. These diseases could be largely preventable by addressing common risk factors, such as unhealthy diet, physical inactivity, tobacco use, and alcohol abuse. However, these risk factors are not randomly distributed in the population, but are influenced by the social determinants of health (SDoH).
SDoH, including socioeconomic status, education, occupation, income, gender, green space, noise, air pollution and so on, are the non-medical factors that influence health outcomes. The SDoH create health inequities among different groups of people, affecting their exposure and vulnerability to CNCDs and their access to prevention and treatment services. For example, people living in poverty may have limited access to healthy food, safe water, health care, and education, which increases the risk of NCDs. Similarly, women may face gender-based discrimination and violence that affect their mental and physical health.
Current studies have found that SDoH are associated with various health outcomes, especially those related to CNCDs, but there is a lack of high-quality cohort data to provide relevant data on environmental monitoring, demographics, and health outcomes to confirm the causal relationship between SDoH and CNCDs. We believe that the potential mechanisms between SDoH and CNCDs are complex. There may be multiple pathophysiological processes and molecular mechanisms involved. We aim to assess the association between SDoH and CNCDs and explore their potential pathways. This study will enhance our understanding of the mechanisms linking SDoH and CNCDs.
We believe genetic variation may explain the relationship between SDoH and CNCDs. Using Mendelian Randomisation, we’ll examine if there’s a causal link. We’ll identify genetic variants linked to SDoH through screening SNPs associated with exposure factors from meta-analyses. Then, we’ll use UK Biobank dataset to investigate if these variants increase CNCD risk. If linked to both SDoH and CNCDs, we can conclude SDoH is a causal risk factor, as genetic variants are randomly allocated at birth and not influenced by behavioural, socioeconomic or physiological factors.
The expected outcomes of this research project are to provide new evidence on how SDoH affect CNCDs; to identify the key SDoH factors that need to be addressed to prevent or reduce CNCDs; and to inform policies and interventions that aim to promote health equity and reduce the burden of CNCDs.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-static-and-dynamic-systemic-inflammatory-markers-and-atherosclerotic-cardiovascular-disease

Association Between Static and Dynamic Systemic Inflammatory Markers and Atherosclerotic Cardiovascular Disease

Last updated:
ID:
587313
Start date:
5 April 2025
Project status:
Current
Principal investigator:
Mr Chang Sheng
Lead institution:
Xiangya Hospital of Central South University, China

Aim: Our study aims to investigate the association between static/dynamic systemic inflammatory markers and atherosclerotic cardiovascular disease (ASCVD). Specifically, systemic inflammatory markers include C-reactive protein (CRP), inflammatory cell counts, and cystatin C. Dynamic systemic inflammatory indices will be derived using trajectory analysis and unsupervised clustering methods. Additionally, we will incorporate exploratory analyses using inflammation indices developed from metabolomics or proteomics data to further characterize systemic inflammation. ASCVD is defined as coronary events (acute myocardial infarction and angina), cerebrovascular events (stroke and transient ischemic attack), and peripheral vascular disease.
Scientific Rationale: Recent studies reveal that more than half of ASCVD patients exhibit systemic inflammation, as evidenced by elevated CRP levels. Alarmingly, patients with higher levels of inflammation demonstrate significantly increased healthcare resource utilization, as well as elevated risks of major adverse cardiovascular events, heart failure, and mortality. Therefore, it is imperative to explore the causal relationship between systemic inflammation and ASCVD.
Project Duration: The study will utilize data from the UK Biobank and is expected to last for three years.
Public Health Impact: The findings will be disseminated through academic publications. In the future, precise monitoring and management of inflammation levels may enable more personalized approaches to cardiovascular disease prevention, ultimately improving patient outcomes and quality of life.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-systemic-immunity-and-cholelithiasis

Association between systemic immunity and cholelithiasis.

Last updated:
ID:
91660
Start date:
14 September 2022
Project status:
Current
Principal investigator:
Dr Bo Zhang
Lead institution:
Fudan University, China

Gallstone disease is a highly prevalent disease with an incidence of 10-20% in the general population. Besides making people feel pain and discomfort, gallstone disease increases social health burden. The disorder of immunity system was linked to gallstone disease but associated studies were not comprehensive.
In this study, we would explore the effect of immunologic factors on gallstone disease. UK Biobank clinical data on gallstone disease, gallbladder operations, and blood sample tests will be collated. We will then compare patients who have gallstones to those who do not, after adjusting for confounders, we would evaluate the role of all kinds of peripheral immune cells in gallstone disease. This will allow us to identify immune system changes which lead to the development of gallstone disease. We will also assess the association between diet, exercise and various environmental factors and the immune system and their impact on the development of gallstones. Hence, we would like the full cohort of patients who have data available on the development of gallstones , blood tests and environmental factors, including diet, exercise and sociodemographic factors.
This study would explore the role of the immune factors in gallstone disease and provided novel insights into the complex pathophysiological mechanisms of human gallstone disease. Modulating immune factors might act as prevention target for gallstone disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-the-triglyceride-glucose-index-and-combined-obesity-indicators-with-chronic-pain

Association between the triglyceride-glucose index and combined obesity indicators with chronic pain

Last updated:
ID:
615363
Start date:
8 April 2025
Project status:
Current
Principal investigator:
Miss Siyi Wang
Lead institution:
Southern Medical University, China

Research questions: triglyceride-glucose (TyG) index is a reliable surrogate for insulin resistance (IR), which is associated with multiple metabolic disorders and cardiovascular diseases. Current research on the relationship between TyG and chronic pain is unclear.
Objectives: The purpose of this study is to investigate the correlation between TyG and its combined obesity indicators (TyG-WHtR, TyG-BMI, TyG-WC) and chronic pain.
Scientific rational: Using data from the UK Biobank, TyG and its combined obesity index (TyG-WHtR, TyG-BMI, TyG-WC) were calculated for each participant according to the formula. Logistic regression, restricted cubic spline (RCS), subgroup analysis, and receiver operating characteristic (ROC) curve were used to analyze the correlation between TyG and combined obesity indicators and chronic pain. Two-sample Mendelian randomization (TSMR) was used to analyze the causal relationship between TyG and combined obesity indicators with chronic pain.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-between-ultraprocessed-food-consumption-and-risk-of-incident-ckd-a-cross-sectional-and-prospective-analysis-in-the-uk-biobank

Association Between Ultraprocessed Food Consumption and Risk of Incident CKD: A Cross-sectional and Prospective Analysis in the UK Biobank

Last updated:
ID:
109828
Start date:
7 September 2023
Project status:
Current
Principal investigator:
Dr Yan Huang
Lead institution:
The Second Affiliated Hospital of South China University of Technology, China

Aims:The aim of our research project is to investigate the relationship between eating highly processed foods and the risk of developing chronic kidney disease (CKD). We want to understand if there is a connection between the consumption of these foods and the likelihood of developing CKD.
Scientific Rationale: We are aware that processed foods have become more common in our diets. These foods tend to be high in calories, unhealthy fats, sugars, and salt, while being low in essential nutrients. Some studies have suggested that eating too many processed foods can lead to various health problems, like obesity, diabetes, and heart disease. However, we don’t know much about the link between processed food consumption and kidney health. Understanding this potential connection is important because it can help guide us towards making healthier food choices and preventing CKD. Our research project will involve analyzing data from the UK Biobank, a large-scale study that follows a large number of people over time. We will look at the participants’ diets and track their kidney health to see if there is an increased risk of CKD among those who consume a lot of processed foods.
Project Duration: This research project is expected to take three years to complete. This timeframe will allow us to thoroughly analyze the data, considering factors that may influence the results, and account for long-term dietary patterns and kidney health outcomes.
Public Health Impact: The findings from this research project can have a significant impact on public health efforts aimed at preventing kidney disease. If we can establish a clear link between eating processed foods and the risk of developing CKD, it can help raise awareness about the importance of diet for kidney health.
In summary, our research project aims to investigate the association between processed food consumption and the risk of developing CKD. By exploring this connection, we hope to provide valuable insights that can inform public health strategies and contribute to preventing CKD.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-beween-cognitive-function-and-chonic-obstructive-pulmonary-disease

Association beween cognitive function and Chonic Obstructive Pulmonary Disease

Last updated:
ID:
1159
Start date:
1 January 2013
Project status:
Closed
Principal investigator:
Dr Daisy Janssen
Lead institution:
CIRO+, Netherlands

Chronic Obstructive Pulmonary Disease(COPD) is a progressive disease, characterized by a gradual decline in lung function. It is a major course of morbidity and mortality worldwide. Patients with COPD often suffer from comorbidities, such as cardiovascular disease, osteoporosis, and psychological symptoms. Previous studies suggest that cognitive functioning might be impaired in patients with COPD and may even predict mortality and disability. However, the exact relationship between lung function and cognitive functioning remains unknown. Results of existing studies concerning this relationship are inconsistent. Several studies suggest that physical activity is associated with the maintenance and improvement in cognitive function in COPD. Further studies are needed to explore this relationship. Understanding of the relationship between lung function, physical activity and cognitive functioning may help to prevent, to recognize and to develop optimal care programs for patients with impairment in lung function and cognitive impairment. Therefore, the aims of this study are: 1) to compare cognitive functioning between patients with and without COPD; 2) to analyze the relationship between severity of impairment in lung function and severity of cognitive impairment; and 3) to study the relationship between cognitive functioning and the level of physical activity in persons with and without COPD. This project requires the use of data only (relating to medical conditions at baseline, cognitive function and physical activity) on the full cohort. This project will help us to understand how cognitive impairment is related to COPD, which may have important consequences for management of the condition.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-accelerometer-derived-sleep-measures-with-liver-and-brain-a-prospective-cohort-study

Association of accelerometer-derived sleep measures with liver and brain: A prospective cohort study

Last updated:
ID:
425620
Start date:
19 April 2025
Project status:
Current
Principal investigator:
Miss Tongwen Liang
Lead institution:
Xi'an Jiaotong University, China

The relationship between sleep disturbances or alterations in sleep patterns and the onset of neurological diseases, such as stroke, brain cancer and cognitive disorder, as well as whether this relationship is influenced by liver disease, remains unclear. The aim of this project is to better understand these associations and provide a scientific basis for the prevention of neurological and cardiovascular diseases. The duration of the project is about three years. We hope to shed light on the effects of sleep on liver and brain health disease and provide a basis for developing effective prevention and intervention strategies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-accelerometer-measured-physical-activity-with-degenerative-musculoskeletal-diseases-a-prospective-study-from-uk-biobank

Association of accelerometer-measured physical activity with degenerative musculoskeletal diseases: a prospective study from UK Biobank

Last updated:
ID:
292298
Start date:
12 December 2024
Project status:
Current
Principal investigator:
Dr Linbo Peng
Lead institution:
Sichuan University, China

DMDs are structural and functional failures of the musculoskeletal system. As the global population ages, DMDs are becoming more prevalent. Mounting evidence supports the benefit of regular PA for improvement of cardiovascular health and reduction in risk of all- cause mortality. However, the association between PA and DMDs has not been established.
We aimed to obtain accelerometer-measured moderate-intensity and vigorous-intensity physical activities and total volume of PA, over a 7-day period in 2013-2015 without prior or concurrent DMDs in the UK Biobank cohort.
We excluded participants who had been diagnosed with DMDs from follow-up hospital records, before the end of their accelerometer wear (prevalent cases). Follow-up time was calculated as person-time in months for each participant from the final date of accelerometer wear to the first occurrence of DMDs or the end of study. The analytic sample consisted of participants who had complete data for PA, age, sex, ethnicity, age completed full time education, Townsend Deprivation Index, smoking, and alcohol consumption.
We used multivariable-adjusted Cox proportional hazards regression models to estimate hazard ratios (HRs) for the association between total volume, moderate-and vigorous PAs, and risk of DMDs.
We assessed the shape of the relationship between moderate-intensity and vigorous-intensity, total volume of PA, and incident DMDs using a restricted cubic spline model. For this purpose, we trimmed observations less than 5% and greater than 95% of the distribution. We specified the knots at the 25th, 50th, and 75th centiles that were used for the categorisation of the variables for total volume, moderate-intensity, and vigorous-intensity of PA.
We plan to establish the association of accelerometer-measured physical activity with degenerative musculoskeletal diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-air-pollution-green-space-lifestyle-and-their-interactions-with-mental-health-and-dementia-a-multinational-comparative-analysis-across-different-populations

Association of air pollution, green space, lifestyle, and their interactions with mental health and dementia: a multinational comparative analysis across different populations

Last updated:
ID:
93921
Start date:
8 November 2022
Project status:
Current
Principal investigator:
Professor Ying Zhou
Lead institution:
Huazhong University of Science and Technology, China

Both air pollution and residential greenness have been shown to affect health, whereas more prospective cohort studies are warranted for causal pathways. Furthermore, how environmental factors and lifestyle behaviours interact to cause or modulate dementia and mental disorder remains unclear. Due to environmental exposure and lifestyle vary by contexts of geography and cultures, this geography-culture-related heterogeneity should be considered in studies of the relationship between environmental exposure and lifestyle with mental health and dementia. Although few comparative studies have been conducted among multicultural populations, the conclusions are mixed and with no inclusion of Asian countries. In recent years, more and more data on lifestyle, environmental exposure, mental health, and dementia outcomes is available in the UK biobank providing an effective way for us to conduct a multinational comparative analysis. In this study, we aim to assess the correlation between environmental exposures (air pollution and residential greenness) and lifestyle (physical activity, dietary pattern, nutritional supplementation, sleep pattern, smoking, and alcohol use) with the health outcomes of dementia and mental disorder using Cox proportional hazards models, adjusted for a range of covariances. We aim to clarify the following questions: what are the different characteristics in the development of depressive disorder and dementia between Asian and European populations; which environmental and lifestyle factors contribute to the differences in the development of depressive disorder and dementia; how environmental factors and lifestyle behaviors interact with each other and whether the potential patterns of synergistic effects between air pollution, greenness, and lifestyle are different in different ethnic populations. The project will be divided into several sub-projects and is expected to last approximately 3 years. To our knowledge, our study is the first multinational comparative study including European and Asian populations to examine the association between environmental exposure and lifestyle with the occurrence of dementia and mental disorder, which will provide a template for future international comparative studies. We hope our multinational study would help provide new directions for green space planning, air pollution control, and better mental disorder and dementia prevention.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-anti-viral-vaccines-apoe-and-genetic-susceptibility-to-herpes-virus-infection-with-incident-dementia

Association of anti-viral vaccines, APOE and genetic susceptibility to herpes-virus infection with incident dementia

Last updated:
ID:
64966
Start date:
14 September 2021
Project status:
Closed
Principal investigator:
Dr Christian Schnier
Lead institution:
Queen's University Belfast, Great Britain

Recent studies have suggested an association of (herpes) virus infection with dementia. This would suggest that vaccination against herpes virus infection (Varicella Zoster Vaccination) might be associated, as well. In our study we will explore if this association can be repeated in the UK Biobank population. We will also study, if the association is effected by the genetic make-up that makes some people more likely to develop dementia, and the gentic make-up that makes people more likely to be effected by herpes viral disease in older ages (shingles). The project will use information from linked routinely collected health data (hospital admission, GP records, mortality statistics) and the genotype information available for the UK Biobank population. We hope to have the data analysed and the results interpreted within an 18 months time period. Our study will help to further understand the suggested association of viral infection with dementia and will help to guide (national) dementia prevention strategies. The project is supported by the Bill Benter Foundation, US.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-antibiotic-exposure-in-early-life-and-cognitive-function-frailty

Association of antibiotic exposure in early life and cognitive function/frailty

Last updated:
ID:
92597
Start date:
8 September 2022
Project status:
Current
Principal investigator:
Dr Zhen Xuemei
Lead institution:
Shandong University, China

Studies show that the composition of the infant’s gut microbiota changes during antibiotic treatment, and increases the risk of developing childhood disease that may persist into adulthood. It has been observed in humans that differences in, or interventions of, the gut microbiota have translated into alterations in cognitive performance. Epidemiological studies have also revealed that early-life antibiotic exposure is associated with worse cognitive outcomes in children, and causes changes in behavior. In addition, rodent studies have shown that administration of high doses of antibiotic has long-term effects on brain neurochemistry and behavior. However, to our knowledge, there are few studies examining long-term/recurrent antibiotic exposure in early life with subsequent cognitive in childhood and elder. In addition, there are no studies investigating the association between long-term/recurrent antibiotic exposure as child/teenager and frailty.
We aimed to examine the association between antibiotic exposure in early life and cognitive function in childhood and elder, whilst considering the roles of gender; and to investigate the association between antibiotic exposure in early life and frailty.
Findings will provide a better understanding of potential influencing of antibiotics throughout life, and will highlight the importance in the rational use of antibiotics for child and teenager.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-beverage-intake-with-all-cause-mortality-and-cardiovascular-outcomes-in-individuals-with-hypertension

Association of beverage intake with all-cause mortality and cardiovascular outcomes in individuals with hypertension

Last updated:
ID:
106321
Start date:
23 June 2023
Project status:
Current
Principal investigator:
Dr Xiang Zhou
Lead institution:
Second Affiliated Hospital of Soochow University, China

Hypertension is the leading risk factor for cardiovascular disease and ranks first as a cause of disability-adjusted life-years worldwide. More than one billion people worldwide suffer from hypertension, which substantially burdens the global health economy. Dietary intervention is recommended to prevent the onset of hypertension and reduce cardiovascular risk by international hypertension guidelines. Numerous studies have shown that moderate alcoholic and nonalcoholic beverage intakes, such as tea, coffee, and red wine, were associated with lower mortality risks and cardiovascular diseases in the general population. Nevertheless, insufficient evidence shows the relationship between beverage intake and cardiovascular diseases in individuals with hypertension. It is, thus, necessary to assess the association of beverage intakes with long-term prognosis in individuals with hypertension. The purpose of our study is to investigate the association between beverage intake and long-term prognosis in individuals with hypertension using a prospective cohort study. Our project duration is expected to be three years. Our study will provide important evidence about the association of beverage intake with cardiovascular disease and death in individuals with hypertension and fill this knowledge gap.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-bone-mineral-density-with-cardiometabolic-disease

Association of Bone Mineral Density with Cardiometabolic Disease

Last updated:
ID:
97015
Start date:
6 July 2023
Project status:
Current
Principal investigator:
Dr Guang Hao
Lead institution:
Guangdong Pharmaceutical University, China

Aims: To study the associations between bone mineral density (BMD) and cardiometabolic risk, and the roles of sun exposure, calcium supplements, and physical activity in those associations.
Scientific rationale: The causes of cardiometabolic disease are complex and still not fully studied.
It is important to identify modifiable risk factors for cardiometabolic disease. Low BMD is linked to cardiometabolic diseases such as type 2 diabetes, myocardial infarction, coronary heart disease, heart failure, and stroke. However, the causal associations are not yet confirmed. Therefore, as a largely preventable condition, low BMD could be a novel target to prevent cardiometabolic disease.
Project duration: 24 months
Public health impact: Our research will clarify the associations between BMD and cardiometabolic disease, and the roles of sun exposure, calcium supplements, and physical activity using this large cohort of UK biobank. The findings may help to build evidence-based strategies to reduce the burden of cardiometabolic disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-brain-structure-and-dementia

Association of Brain Structure and Dementia

Last updated:
ID:
45449
Start date:
3 July 2019
Project status:
Closed
Principal investigator:
Dr Narges Razavian
Lead institution:
NYU Grossman School of Medicine, United States of America

Alzheimer’s disease is common, terminal and does not have a cure at the moment. By the time clinical symptoms emerge, it is years too late to be able to reverse the disease. 100% of clinical trials have failed so far, partly because they recruit patients when they already have clinical symptoms.
Our brain-changes start *decades* before any clinical symptom of AD emerges. Currently, there are no accurate imaging biomarkers from MRI that would allow us to detect the disease at its earlier stage. Existing methods of today are expensive, unpleasant to the patient, and involve injection of radioactive markers(PET), something we can not do at regular intervals. Therefore we are not very good at detecting AD before clinical symptoms start.
On the other hand, artificial intelligence has shown great success at sifting through thousands of images, and finding patterns that differentiate objects from each other. Our project aims to use AI to look into thousands of brain MRI scans, find patterns that are associated with early and late Alzheimer’s disease stage, compared to normal aging. If completed, this can impact clinical trials and development of treatments for a disease that has evaded cure up to now.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-cardiovascular-health-assessed-by-the-lifestyle-factors-metrics-with-common-non-communicable-diseases

Association of Cardiovascular Health Assessed by the Lifestyle Factors Metrics With Common Non-Communicable Diseases

Last updated:
ID:
293943
Start date:
27 November 2024
Project status:
Current
Principal investigator:
Professor Yilan Li
Lead institution:
Harbin Medical University, China

Aims: The aim of this study is to evaluate the association between cardiovascular health indicators and common non-communicable diseases. Deeply explore the mutual influence and correlation between cardiovascular health, genetic environmental factors, and common non-communicable diseases, and analyze the potential regulatory role of cardiovascular health in the relationship between environmental pollution exposure/genetic risk and common non-communicable diseases.

Scientific Rationale:
Many diseases stem from the mutual influence of genetic, environmental, and lifestyle risk factors. According to data from the World Health Organization, the impact of lifestyle on health and lifespan accounts for 60%, genetic factors account for 15%, environmental and social factors account for 17%, and medical conditions account for 8%. Nowadays, more and more people suffer from a variety of non communicable diseases, such as diabetes, cancer, cardiovascular disease and chronic obstructive pulmonary disease. However, the potential interaction between cardiovascular health indicators, genetic factors, and environmental factors on chronic diseases has not been fully studied. Further research is needed to determine whether (and to what extent) cardiovascular health can offset the risk of chronic diseases caused by genetic susceptibility or environmental exposure.

Project duration: This project is expected to be completed in 36 months.

Public Health Impact: The primary goal is to validate the effectiveness of cardiovascular health indicators in predicting and reducing the incidence of chronic non-communicable diseases. Additionally, the project aims to uncover the complex interactions between cardiovascular health, genetic risks, and environmental factors, enhancing public awareness and adherence to these health indicators to mitigate chronic disease burdens.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-cardiovascular-health-cvh-and-healthy-lifestyle-index-hli-with-the-incidence-of-and-mortality-from-major-chronic-diseases

Association of Cardiovascular Health (CVH) and Healthy Lifestyle Index (HLI) with the incidence of and mortality from major chronic diseases.

Last updated:
ID:
107335
Start date:
4 September 2023
Project status:
Current
Principal investigator:
Dr Chuang Yang
Lead institution:
University of Leipzig, Germany

Scientific rationale:
Diversity of lifestyle factors including such as cigarette smoking, body mass index, diet, physical activity, total cholesterol, fasting plasma glucose and blood pressure, waist circumference, alternative healthy eating Index and alcohol consumption result a deeply effect on the risk of getting chronic diseases, and may also be linked to chances of surviving after disease diagnosis. Lifestyle factor indices such as cardiovascular health (CVH) and healthy lifestyle index (HLI) have positive preventive implications in assessing the risk of chronic diseases, due to the fact that people’s lifestyles can be adjusted on their own. Previous studies also showed a strong association between CVH and HLI and the mortality of patients. However, studies with large samples have not been widely reported. Furthermore, it is important to understand how these lifestyle indices behave in different populations and to compare which index better predicts incidence or mortality from chronic diseases.

Aims:
This study aims to investigate the association of CVH and HLI with the incidence of and mortality from major chronic diseases. Major chronic diseases included cardiovascular disease, diabetes mellitus, dementia, and cancer. We will also focus on analyzing how CVH and HLI relate to different cardiovascular disease and cancer phenotypes, which will be of great value in gaining a deeper understanding and preventing these diseases. This research direction is in line with the trend towards precision medicine.

Project duration:
Over a three-year project, we will use data on participant characteristics, lifestyle (cigarette smoking and alcohol consumption, body mass index, diet, physical activity, lipid levels, fasting plasma glucose and blood pressure, waist circumference, alternative healthy eating Index and hours of night sleep) from the UK Biobank Study. Moreover, we will select reasonable participants and obtain CVH and HLI scores, and examine the relationship between CVH and HLI and the incidence and mortality of four major chronic diseases.

Public Health Impact:
The findings from this study could help to guide lifestyle recommendations and public health policies for the general population, and for those suffering from diseases, to reduce the risk of chronic diseases and improve length, and quality, of life. By exploring the utility and quality of scoring systems and indices used to measure lifestyle ‘healthfulness’, this research could provide more accurate tools to be used in future studies worldwide.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-cigarette-smoking-patterns-with-all-cause-and-cause-specific-mortality-in-patients-with-prostate-cancer

Association of cigarette smoking patterns with all-cause and cause-specific mortality in patients with prostate cancer

Last updated:
ID:
493929
Start date:
3 January 2025
Project status:
Current
Principal investigator:
Dr Xiangwei Yang
Lead institution:
The Seventh Affiliated Hospital of Sun Yat sen University, China

The association between cigarette smoking and the risk of prostate cancer remains controversial. Cigarette smoking at the time of diagnosis has been linked to increased risks of biochemical recurrence, metastasis, and mortality among patients with prostate cancer. However, post-diagnosis smoking patterns in patients with prostate cancer are rarely reported, and the impact of continued smoking and smoking cessation on the prognosis of prostate cancer remains unclear. We perform this project to investigate the patterns and correlates of cigarette smoking in patients with prostate cancer and examine the association of cigarette smoking patterns after cancer diagnosis with all-cause and cause-specific mortality.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-clonal-haematopoiesis-of-indeterminate-potential-mutations-with-clinical-complications

Association of clonal haematopoiesis of indeterminate potential mutations with clinical complications

Last updated:
ID:
771460
Start date:
26 June 2025
Project status:
Current
Principal investigator:
Professor Anskar Yu Hung Leung
Lead institution:
Centre for Oncology and Immunology Limited, Hong Kong

Clonal haematopoiesis of indeterminate potential (CHIP) refers to the phenomenon in which mutations occur in ageing haematopoietic stem cells (HSCs), resulting in clonal selection and expansion. CHIP was defined by the presence of white cells in peripheral blood carrying the characteristic CHIP mutations with a variant allele fraction (VAF) greater than 2%. It is now becoming clear that CHIP is associated with a pro- inflammatory state and increased risks for haematological malignancies and cardiovascular and cerebrovascular diseases (1, 2). Reportedly, the prevalence of CHIP mutation increases with age, occurring in 10% of people over 70 years old and 20% of those over 90 years old (3). Furthermore, it is unclear how specific CHIP mutations, singly or in combination, are associated with specific complications. Such information may form the foundation for Personalized Disease Prevention (PDP) for the ageing population.

The purpose of this project is to find the association of clonal haematopoiesis of indeterminate potential (CHIP) mutations with specific clinical complications based on genetic and clinical data derived from UK Biobank.

Specific objectives of the project are:
1) Evaluate association of specific CHIP mutations with clinical phenotypes.
2) Examine how the presence of two or more CHIP mutations affect clinical phenotypes.

References:
1. Jaiswal S, Natarajan P, Silver AJ, Gibson CJ, Bick AG, Shvartz E, et al. Clonal Hematopoiesis and Risk of
Atherosclerotic Cardiovascular Disease. N Engl J Med 2017 Jul 13; 377(2): 111-121.
2. Xie M, Lu C, Wang J, McLellan MD, Johnson KJ, Wendl MC, et al. Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nat Med 2014 Dec; 20(12): 1472-1478.
3. Jaiswal S, Fontanillas P, Flannick J, Manning A, Grauman PV, Mar BG, et al. Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med 2014 Dec 25; 371(26): 2488-2498.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-combined-exposures-to-greenness-and-air-pollution-with-cardiovascular-diseases-biomarkers

Association of combined exposures to greenness and air pollution with cardiovascular diseases biomarkers

Last updated:
ID:
65036
Start date:
15 February 2021
Project status:
Current
Principal investigator:
Dr John S. Ji
Lead institution:
Tsinghua University, China

Cardiovascular disease (CVD) is the world’s leading cause of death. The protective effect of greenness on CVD is becoming more evident. However, the link between green space and cardiovascular health remains weak, in particular, because it is unclear how exposure to green space influences cardiovascular health and which pathophysiological processes and mechanisms mediate the relationship between green spaces and CVD risk. There is a lack of objective evaluation of the biological processes related to greenness and cardiovascular disease. Meanwhile, air pollutants are acknowledged risk factors of CVD and can be reduced by greenness. We aim to evaluate the associations of combined exposures to greenness and air pollution with cardiovascular disease biomarkers and explore the underlying biologic pathways. With the detailed information about individual environmental exposure, this project will improve our knowledge of the mechanisms linking greenness and cardiovascular disease and the role of air pollution in greenness protection of cardiovascular disease. The information generated from this project will be useful for government and public health policymakers (inform how greenness should be included as part of urban planning), as well as the general public themselves (understanding risks and how to reduce them). The project is expected to last 36 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-currently-and-lifetime-psychological-issues-with-cancer-specific-morbidity-and-mortality

Association of currently and lifetime psychological issues with cancer-specific morbidity and mortality

Last updated:
ID:
96414
Start date:
22 November 2022
Project status:
Current
Principal investigator:
Professor Ji-Bin Li
Lead institution:
Sun Yat-sen University Cancer Center, China

Psychological issues (e.g., depression, insomnia, anxiety, etc.) have become increasingly common in modern society and are associated with increased disease burdens worldwide, and several studies have suggested a positive association between psychological disorders (e.g., depression, insomnia) and the risk of several diseases. psychological problems such as depression could impact the immune system, the endocrine system, cancer metastasis, treatment tolerance, and other processes. The coexistence of multiple psychological disorders (i.e., depression, insomnia, anxiety) are very common among general population. However, it is still unclear causation and the extent to which comorbidity of common psychological issues (i.e., depression, anxiety, insomnia, adolescent and adult Traumatic events) could increase the risk of cancers and mortality. Therefore, we would like to conduct a population-based cohort study to investigate the association of psychological issues (i.e., depression, anxiety, insomnia, adolescent and adult traumatic events) with cancer-specific morbidity and mortality. The study will last for 36 months. Our study will provide the robust results on the association between psychological disorder and all-cause mortality and incidence of and mortality from all cancer, and subtypes of cancer. The findings will be helpful for future primary prevention and intervention among community-based population.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-daily-steps-and-intensity-with-cardiovascular-events-stroke-and-mortality-in-patients-with-chronic-kidney-disease

Association of daily steps and intensity with cardiovascular events, stroke, and mortality in patients with chronic kidney disease

Last updated:
ID:
106309
Start date:
15 November 2023
Project status:
Current
Principal investigator:
Dr Xiu Hong Yang
Lead institution:
Fudan University, China

The risk for cardiovascular events are significantly higher in patients with chronic kidney disease (CKD), lifestyle intervention is important in the prevention of both cardiovascular complications and progression of kidney disease. Daily steps or walking is simple and easy as compared with other physical exercises. This study is to explore the appropriate daily steps to reduce cardiovascular disease (CVD) complications and delay the progression of renal insufficiency using the prospective study data from the UK Biobank cohort. The primary outcome was the occurrence of CVD events, stroke, and cardiovascular-specific mortality and all-cause mortality in patients with CKD. The secondary outcomes are the occurrence of kidney hard endpoints ((creatinine doubling, end-stage renal failure, dialysis, or transplantation), also including the changes of estimated glomerular filtration rate (eGFR) and proteinura. The results will be helpful in the development of future guidelines on exercise for patients with CKD.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-depressive-symptoms-with-risk-of-all-cause-mortality-cause-specific-mortality-and-morbidity

Association of depressive symptoms with risk of all-cause mortality, cause-specific mortality and morbidity.

Last updated:
ID:
63871
Start date:
17 August 2020
Project status:
Closed
Principal investigator:
Dr Lisa Pennells
Lead institution:
University of Cambridge, Great Britain

We seek to use the unprecedented power and richness of data in UK Biobank to investigate the relationship between depressive symptoms and risk of multiple chronic diseases.

More than 264 million people worldwide are affected by depression, and this burden has increased over the past 10 years. Chronic diseases are the leading cause of death and among the top causes globally of premature years of life lost. Links between chronic diseases and depressive disorders have been recognized for more than 300 years with numerous studies reporting increased risk of chronic diseases in individuals with depressive disorders. However, limitations of previous studies include i) involvement of a small numbers of participants and ii) insufficient examination of the influence of other known risk factors. Furthermore, few past studies have used genetic information to assess the causal nature of any association.
We plan to use data in UK Biobank to better quantify the observed association between depressive symptoms and risk of multiple chronic diseases. Assuming an association is observed, we also aim to use the genetic information available in UKBiobank participants to help determine whether the nature of this association is causal, by examining whether individuals with genetic predisposition to elevated depressive symptoms are also at increased risk of multiple chronic diseases.

The specific research objectives are:

(1) To characterise and compare the associations of depressive symptoms with future risk of multiple chronic diseases.

(2) To examine the relationship between depressive symptoms and other risk factors, and check whether any of these other risk factors impact on the observed relationship between depressive symptoms and multiple chronic diseases.

(3) To provide evidence for any causal association between depressive symptoms and disease, thereby eliminating other potential reasons for the association: i.e. that risk is being effected by other co-occurring risk factors or that an individual’s experience of depressive symptoms may to be linked to underlying unknown diseases (a phenomenon known as reverse causality).

Results can advance understanding of factors related to levels of depressive symptoms and clarify the relative importance of depressive symptoms on disease risk. This, in turn, will inform future research which could potentially impact on clinical practice to reduce burden of both depression and chronic diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-device-measured-physical-activity-with-cardiovascular-disease-and-death-in-individuals-with-hypertension

Association of device-measured physical activity with cardiovascular disease and death in individuals with hypertension

Last updated:
ID:
98764
Start date:
9 March 2023
Project status:
Current
Principal investigator:
Dr Xiang Zhou
Lead institution:
Second Affiliated Hospital of Soochow University, China

Hypertension is defined as office systolic blood pressure values >=140 mmHg and/or diastolic blood pressure values >=90 mmHg, strongly associated with cardiovascular diseases and death. Over one billion people worldwide suffer from hypertension, and it is expected to reach 1.5 billion by 2025, which places a substantial burden on the global health economy. Tremendous studies have shown that increased physical activity (PA) can effectively lower blood pressure. A narrative review of 27 randomized clinical trials showed that regular moderate-to-vigorous intensity PA reduced blood pressure by approximately 11/5 mmHg in individuals with hypertension. The 2020 World Health Organization (WHO) guideline on PA recommends at least 150-300 minutes of moderate-intensity PA or 75-150 minutes of vigorous-intensity PA or an equivalent combination throughout the week to reduce the risk of cardiovascular diseases and death. Nevertheless, few studies demonstrated that this PA recommendation effectively improves cardiovascular prognosis in the population with hypertension. It is still being determined whether the blood pressure reductions related to physical activity translate into long-term benefits. Our study aims to investigate the association of physical activity with cardiovascular disease and death in individuals with hypertension by conducting a prospective cohort study. Our project duration is expected to be three years. Our study will provide important evidence about the association of physical activity with cardiovascular disease and death in individuals with hypertension and propose some physical activity advice for patients with hypertension.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-diabetes-and-its-complications-with-chronical-diseases

Association of diabetes and its complications with chronical diseases

Last updated:
ID:
95552
Start date:
6 December 2022
Project status:
Current
Principal investigator:
Dr Yong Chen
Lead institution:
Anhui Medical University, China

The project aims to study the potential relationships between diabetes and its complications and common chronic disorders (such as hypertension, COPD, CVD, and osteoporosis). Whether hyperglycemia has any influence on chronical. Do people with diabetes report more likely to chronical diseases, compared to those without diabetes?

Type 2 diabetes is a common disease that is caused by a combination of multiple genetic, environmental and behavioral factors. In order to identify the relationship of diabetes and chronical diseases, it is necessary to explore the long-term hyperglycemia effects on the development and outcomes of chronical diseases from the perspective of population charasteristics, genetic, biomarkers.

The duration of this project is approximately 3 years. The results of this project will reveal the potential associations between diabetes and its complications and chronic diseases. Therefore, these findings could tell us about possible causes or factors of the development and outcomes of chronical disease, thereby providing more information for clinicians and therapists to prevent the happening and decrease the risk of death from these disorders.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-diabetes-mellitus-with-spinal-degenerative-disorders-an-observational-and-two-sample-mendelian-randomization-study

Association of diabetes mellitus with spinal degenerative disorders: an observational and two-sample mendelian randomization study

Last updated:
ID:
91061
Start date:
9 August 2022
Project status:
Current
Principal investigator:
Professor Jiang-Hua Liu
Lead institution:
University of South China, China

Aims: The purpose of the current project is to clarify the causal correlation between diabetes mellitus (DM) and spinal degenerative disorders (SDD).

Scientific rationale: DM and SDD are two commonly encountered pathologies in clinical practice. DM is a chronic systematic disease that can involve multiple organs, while SDD represents an aging-related musculoskeletal disease that is caused by intervertebral disc degeneration (IDD). It has been reported that DM can affect nearly all connective tissues, including bone and cartilage. In support of this, published data have demonstrated that DM is implicated in the development and deterioration of osteoarthritis. As an analogical condition to osteoarthritis, however, whether SDD can also be caused or promoted by DM remains to be fully elucidated. Previous clinical and laboratory studies have indicated that DM is a contributing factor to intervertebral disc degeneration, although this finding is still controversial. Given the fact that disc degeneration is an initiating pathological process for SDD, we therefore hypothesize that DM may also likely be a culprit of SDD.

Expected duration of project: The estimated duration of this project is three years.

Public health impact: Our project will clarify the causal effect of DM on SDD risk, which can be useful for risk stratification and therapeutic optimization of SDD in DM patients.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-dysanapsis-genetic-risk-with-lung-function-and-health-outcomes

Association of dysanapsis genetic risk with lung function and health outcomes

Last updated:
ID:
97748
Start date:
31 January 2023
Project status:
Current
Principal investigator:
Dr Benjamin Smith
Lead institution:
Research Institute McGill University Health Centre, Canada

Chronic obstructive lung diseases represent a major cause of death and disability in the UK and globally.
Tobacco smoke is the best known risk factor, but only a minority of lifelong tobacco smokers develop chronic obstructive lung disease, suggesting that other factors must contribute.
Dysanapsis refers to a developmental mismatch between airway tree and lung size. Dysanapsis assessed by computed tomography (CT) scan has been shown to be established by early adulthood, and is associated with much higher risk of chronic obstructive lung disease later in life. A recent genetic study identified several polymorphisms associated with dysanapsis and, when combined into a dysanapsis genetic risk score, children with higher dysanapsis genetic risk showed abnormal lung function at 5 years old. This study seeks to test the following hypotheses among UKBiobank participants:
1 Higher dysanapsis genetic risk is associated with low lung function
2 Higher dysanapsis genetic risk is associated with higher rates of death and hospitalization
3 Higher dysanapsis genetic risk increases susceptibility to tobacco smoke and air pollution


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-education-level-with-all-cause-and-cause-specific-mortality-among-individuals-with-t2dma-prospective-study-in-uk-biobank

Association of education level with all cause and cause-specific mortality among individuals with T2DM:a prospective study in UK Biobank

Last updated:
ID:
95356
Start date:
22 November 2022
Project status:
Current
Principal investigator:
Dr Yan Liu
Lead institution:
Dongzhimen Hospital, Beijing University of Chinese Medicine, China

Diebetes is more and more epidemic worldwide.Diabetes can not noly elevate our blood gluose but also lead to some complications which will shorten our lifetime, decrease quality of our life and add our financial burden.Which factor is linked to the diabetes? We want to explore whether diabetic health conditions will be affected by education level.The reasons for doing this research are as follows.Firstly,according to our survey,we have kown that diabetes is influenced by our diet,physical activity and other lifestyle.Many social factors can be reflected by education level such as income, occupation,social status and so on.From educational level, we can have a comprehensive understanding of patient’s conditions.Secondly,educational level refers to our ability to learn knowledge like health literacy. The more educated the public is ,the more concerned they are about health problems , and the better management they will have of diabetes, thus control or delay the development of diabetes.The evidence on educational level is limited.To fill the knowledge gap,we plan to evaluate diabetes educational level from UK biobank and find an objective outcome .Our aim is to help the public know more about diabetics and can get benifit from it.The estimated duration of our project is about 36 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-egg-consumption-with-the-risk-and-prognosis-of-dementia-in-older-people

Association of egg consumption with the risk and prognosis of dementia in older people

Last updated:
ID:
80300
Start date:
21 December 2022
Project status:
Current
Principal investigator:
Dr Precious Osayuki Igbinigie
Lead institution:
University of Wolverhampton, Great Britain

Owing to the increase cases of dementia as a result of the early onset of the disease and the increased life expectancy there is a need for an effective approach to reduce the number of individual affected by the disease.
Nutritional intervention can be one of the strategic approaches to prevent and manage dementia due to lack of pharmacological intervention to treat the condition. the aim of carrying out this research is to determine how egg consumption influences dementia. Egg being a staple food is commonly consumed by the population owing to his reduced cost and diverse way of preparation. This research will assist health professional in tackling the impact of dementia on the economy as a result of reduced manpower and financial constraints occasioned by the dementia.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-emerging-cardiovascular-disease-biomarkers-and-markers-of-metabolic-health-with-subclinical-atherosclerosis-clinical-atherosclerotic-cardiovascular-disease-and-mortality

Association of emerging cardiovascular disease biomarkers and markers of metabolic health with subclinical atherosclerosis, clinical atherosclerotic cardiovascular disease, and mortality

Last updated:
ID:
97439
Start date:
5 April 2023
Project status:
Current
Principal investigator:
Professor Michael D. Shapiro
Lead institution:
Wake Forest School of Medicine, United States of America

Heart disease is the leading cause of death in the world. There are many causes of heart disease, but most commonly, it is due to atherosclerosis. Atherosclerotic cardiovascular disease is due to damage and blockages of blood vessels that supply oxygen to the heart. Most commonly, individuals with atherosclerosis will go through a long latent (subclinical) period without experiencing a clinical event, such as a heart attack or stroke. It is well known that low-density lipoprotein-cholesterol (LDL-C), or “bad cholesterol”, leads to atherosclerosis. Thus, measuring LDL-C helps to predict cardiovascular risk, and serves as a target for cholesterol lowering drugs. However, there are many other markers that are being studied that could provide further insight in the development of atherosclerosis and other forms of cardiovascular disease. We are interested in looking at how these markers of metabolic and cardiovascular risk relate to one another, as well as the development of subclinical and clinical cardiovascular disease. By doing so, we hope to improve our knowledge regarding the relationship between these various markers and the ability of these markers to help predict subclinical and clinical cardiovascular disease, beyond LDL-C.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-environmental-and-genetic-factors-with-mental-illnesses-through-imaging-genetics

Association of environmental and genetic factors with mental illnesses through imaging genetics

Last updated:
ID:
104766
Start date:
28 February 2024
Project status:
Current
Principal investigator:
Professor Jinsong Tang
Lead institution:
Zhejiang University, China

Aims:Current research has found that conventional lifestyle factors (such as diet, physical activity, smoking and alcohol, electronic devices use), adverse life experience, other individual behaviors (such as social media exposure), and health and medical history have been associated with mental illnesses including psychiatric disorders and cognitive dysfunction. Whether these environmental risk factors play a synergistic role in the genetic mechanism of mental illnesses warrants further investigation. Although abdundant neuroimaging studies have been conducted to look for unique brain structural and functional changes related to mental illnesses, large-sample studies are highly encouraged to estabilish replicatable and solid neuroimaging phenotypes. To sum up, we plan to utilize UK Biobank data to quantify environmental, genetic, and neuroimaging contribution to mental illnesses and establish artificial intelligence-based representation model by means of imaging genetics.
In the time span of 36 months, the aims of this research are: (1) to investigate environmental factors (such as adverse life experience, lifestyle, and psychosocial factors) of mental illnesses including psychiatric disorders and cognitive dysfunction; (2) to identify genetic endotypes of mental illnesses and resultant metabolic disturbances (such as endocrine disorders and cardiovascular diseases); (3) to study gene-environmental interaction of the above-mentioned illnesses; (4) to identify distinct brain structural features by measuring T1 structural brain MRI, T2-weighted brain MRI and diffusion brain MRI, and functional features via resting-state fMRI or arterial spin labelling brain MRI of common mental illnesses; (5) to identify how genetic and environmental risk, through transcriptome regulation, is linked to neural dysconnectivity and how their connection in turn contributes to clinical deficits in mental illnesses; (6) with machine learning methods to establish classification and prognosis prediction models with genetic, environmental, and neuroimaging markers.
We hope the results of this study could help us better understand the onset and development of mental illnesses in a biopsychosocial way and learn more about how genetic and environmental factors contribute to clinical deficits thourgh neural activities.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-environmental-factors-genetic-risk-and-their-interactions-with-cardiometabolic-disease

Association of environmental factors, genetic risk and their interactions with cardiometabolic disease

Last updated:
ID:
478081
Start date:
24 March 2025
Project status:
Current
Principal investigator:
Dr Cheng Long
Lead institution:
Fudan University, China

Cardiometabolic diseases are the predominant cause of mortality, morbidity and healthcare spending globally, and are believed to result in part from the combined additive and synergistic effects of genetic and environmental risk factors. Environmental exposures such as diet and physical activity have enormous potential for prevention and treatment of these diseases, but no single therapy works well in all individuals. Therefore, we raise the question whether there are undiscovered risk factors related to cardiovascular metabolic diseases, if there are, whether there is interaction between these risk factors, and whether they are genetically related.
we want to utilize both quantitative and qualitative data to study the biological, physical or imaging measurements, behavioral, environmental, and genetic factors affecting or predicating the incidence of cardiometabolic diseases, and the risk of adverse outcomes in patients with cardiometabolic diseases. At the same time, we will investigate environmental-genetic interactions on cardiometabolic diseases. We also aim to identify new genetic risk loci or study on the previously identified risk sites of cardiac metabolic diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-environmental-factors-with-chronic-kidney-disease

Association of environmental factors with chronic kidney disease

Last updated:
ID:
226010
Start date:
8 October 2024
Project status:
Current
Principal investigator:
Dr Yang Li
Lead institution:
Haikou Hospital Affiliated to Xiangya Medical College of Central South University, China

Chronic kidney disease (CKD) has become a major public health problem that affects human health worldwide. Despite significant improvements in treatment of CKD in recent decades, patients still have a significant risk of progressive loss of renal function, which can have an negative effect on patients’ quality of life. Previous research has shown that environmental factors result a deeply effect on kidney health. For example, there exists a positive correlation between PM2.5 and the prevalence of kidney disease. However, the current research on the relationship between CKD and environmental factors is not sufficient, so further studies are imperative to comprehensively comprehend and accurately explain this relationship. We aim to investigate the association of environmental factors with chronic kidney disease (CKD) and identify risk factors that are highly associated with the onset, progression, and prognosis of CKD. Our project is estimated to take 36 months to complete. The results of this project will deepen the understanding of the relationship between CKD and environmental factors, and help people understand how environmental factors affect their health so that they can take preventive measures in time. We hope our research will inform public health, environmental science and policy making.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-environmental-non-environmental-factors-and-genetic-risk-scores-with-chronic-noncommunicable-diseases

Association of environmental, non-environmental factors and genetic risk scores with chronic noncommunicable diseases

Last updated:
ID:
150464
Start date:
13 December 2023
Project status:
Current
Principal investigator:
Dr Yuqing Huang
Lead institution:
Guangdong Provincial People's Hospital, China

This research intends to investigate the many factors associated with chronic noncommunicable diseases.These project will include both hereditary and non-genetic variables. It is generally acceoted that many chronic noncommunicable diseases are caused by genetic factors, such as gene variations and associated biomarkers. Furthermore, non-genetic variables are piqueing the interest of researchers due to their role in the development of chronic noncommunicable diseases. Dietary and environmental factors, for example, are common non-genetic contributors. Understanding the underlying causal links between these factors and chronic noncommunicable diseases will have a significant impact on human health and longevity. This project is expected to continue three years or until the objectives are met. Our findings may aid in the identification of biological and environmental components that are linked to a number of chronic noncommunicable diseases. It also aids in the provision of scientific recommendations for medical services, environmental science, and policy formulation.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-environmental-pollution-with-bone-loss-over-time-osteoporosis-and-bone-fracture-risk

Association of environmental pollution with bone loss over time, osteoporosis and bone fracture risk.

Last updated:
ID:
59186
Start date:
25 August 2020
Project status:
Closed
Principal investigator:
Professor Cuiqing Liu
Lead institution:
Zhejiang Chinese Medical University, China

Emerging evidence indicated associations between air pollution exposure and adverse cardiovascular and respiratory health effects. However, associations between environmental pollution (nitrogen dioxide, particulate matter air pollution or road traffic noise) and bone loss, osteoporosis and fracture are not well understood. Research is therefore required to further investigate the effects of environmental pollution (if any) on skeletal health. Identification of risk and protective factors is important to both science and policy decision given the high health care costs related to osteoporosis and fracture morbidity.
Aims:
1 To determine whether exposure to environmental pollution (nitrogen dioxide, particulate matter air pollution or road traffic noise) is associated with bone loss, and/or increased odds of having an osteoporosis or fracture.
2 To determine whether the observed associations from Aim 1 vary on other factors including gender, race/ethnicity, socioeconomic status, age, BMI, smoking status, and others.
3 To identify BMD in different sites (heel, arm, femur and others) to examine the susceptibility of different bone sites to the exposure of environmental pollution.
4 To identify osteoporosis or fracture that might be associated with exposure to environmental pollution.
5 To identify the vulnerable populations (age, sex, ethnicity and others) of osteoporosis or fracture to air pollution exposure.
The estimated project duration is 2 years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-frailty-and-risk-of-colorectal-cancer-in-inflammatory-bowel-disease-patients-a-mendelian-randomization-analysis-findings-from-uk-biobank

Association of Frailty and Risk of Colorectal Cancer in Inflammatory Bowel Disease Patients: A Mendelian randomization analysis/ Findings from UK Biobank

Last updated:
ID:
679066
Start date:
28 September 2025
Project status:
Current
Principal investigator:
Miss Zhu Deng
Lead institution:
Beijing University of Chinese Medicine, China

Research Questions: Does frailty increase the risk of progression from inflammatory bowel disease (IBD) to colorectal cancer (CRC)? Which biological mechanisms mediate the relationship between frailty and the carcinogenic process in IBD? What is the genetic correlation and potential causal relationship between frailty and colorectal cancer?
Objectives: First, to assess the impact of frailty on the progression of IBD to CRC. Secondly, to investigate the mechanisms linking frailty to IBD-related carcinogenesis using data from the UK Biobank (UKB). Thirdly, to analyze the genetic correlation and establish potential causal links between frailty and CRC through Mendelian Randomization (MR).
Scientific rationale:
Global cancer statistics reveal nearly 20 million new cancer cases and 9.7 million cancer-related deaths in 2022. Frailty, a state of heightened vulnerability to stressors, has been linked to adverse health outcomes. Previous studies using UKB data have shown that frail individuals face a higher risk of developing cancer, with frailty identified as a predictor of cancers such as lung cancer.
IBD relapsing inflammation of the gastrointestinal tract due to an aberrant immune response to the gut microbiome. The prevalence of IBD exceeds 0.3% in many developed regions. A well-established association exists between IBD and CRC, UKB data reveal that IBD patients face an adjusted hazard ratio (HR) of 1.54 for CRC compared to the general population. The “inflammation-cancer transformation” model, originally proposed by Correa P. for gastric cancer. Similarly, IBD-associated CRC follows a pathway of “colonic inflammation!dysplasia!carcinoma”. Effective management of IBD is essential to improving patient outcomes and preventing this progression. Despite evidence suggesting that frailty influences cancer development, its role in the transition from IBD to CRC remains underexplored.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-fundus-imaging-findings-with-risk-of-dementia-and-premature-death

Association of Fundus Imaging Findings with Risk of Dementia and Premature Death

Last updated:
ID:
59418
Start date:
4 May 2020
Project status:
Current
Principal investigator:
Dr Nambi Nallasamy
Lead institution:
University of Michigan, United States of America

It is well known that people age differently. Promoting healthy longevity requires that we learn from those who age well, and that we slow the aging process in those who seemingly age too quickly. However, to do so it is necessary to be able to understand which patients are at risk of aging poorly (through the development of dementia or through premature death). The human retina provides a unique window into the health of a human being, as it allows us to directly view components of the central nervous system and cardiovascular system at the same time through non-invasive photographs.

Deep learning is a powerful tool that uses the predictive power of all features contained in an image, and is not restricted to those characteristics that researchers can see and presume to be relevant. Prior research has found that deep learning can be applied to retinal images to identify many non-ocular health traits. In this study, using separate training and validation sets of retinal images from individuals in the UK Biobank, we will develop and test deep learning algorithms to predict risk of dementia and premature death.

This research may catalyze critical advances in biogerontology and medicine. For example, accurate identification of risk of dementia and premature death may be used to measure the efficacy of interventions that aim to slow the aging process; to identify resilience factors in those who age slowly; and to improve scientific understanding of the cellular and molecular changes that drive physiologic aging. Consequently, this project holds the potential to transform the study of healthy longevity.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-genetic-and-lifestyle-factors-with-the-risk-of-intracranial-aneurysm

Association of genetic and lifestyle factors with the risk of intracranial aneurysm.

Last updated:
ID:
78223
Start date:
20 May 2022
Project status:
Current
Principal investigator:
Professor Wanyang Liu
Lead institution:
China Medical University P.R.C, China

As aneurysmal rupture is the main event leading to the occurrence of subarachnoid hemorrhage (SAH) and it often results in high disability or death rates, the management unruptured intracranial aneurysm (UIA) for preventing ruptured intracranial aneurysms (RIA) is crucial. In terms of non-genetic factors, most of the existing studies investigated the influence of some non-genetic factors, such as lifestyle, on intracranial aneurysms (IAs). The influence of non-genetic factors on the occurrence and development of intracranial aneurysm (IA) under different genetic risk (GR) is still unclear.
Here, we expect to conduct this study in three years, after obtaining permission from the UK-Biobank Steering Committee. Our aim is to construct an IA-polygenic risk scores(IA-PRS) for IAs using the results of the currently published genome-wide association study (GWAS) analysis and UK Biobank which was categorized as three levels: low (quintile 1), intermediate (quintiles 2-4), or high (quintile 5). The group with ideal lifestyle and low GR (quintile 1 of IA-PRS) was compared to explore whether GR changes the risk of occurrence and development of IA and whether lifestyle under different GR will change the risk of genetic susceptibility of IAs.
This study is based on the patients with IA among the 500,000 participants of UK Biobank, focusing on their GR and lifestyle, in order to carry out intervention measures as soon as possible in the process of disease occurrence and development, reduce the incidence of IA as much as possible and improve the overall quality of life of patients with IAs.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-genetic-metabolic-and-environmental-risk-factors-with-neuropsychiatric-disorders

Association of genetic, metabolic, and environmental risk factors with neuropsychiatric disorders

Last updated:
ID:
123008
Start date:
16 November 2023
Project status:
Current
Principal investigator:
Professor Yu-Xiang Yan
Lead institution:
Capital Medical University, China

Aims: This study aims to ascertain the causal association between genetic, environmental and metabolic factors and the onset and prognosis of prevalent neuropsychiatric disorders in middle-aged and elderly individuals in Asia and Europe. The objective is to explore the joint effects of genetic, metabolic, and environmental risk factors on neuropsychiatric disorders.
Sientific rationable: Neuropsychiatric disorders (including dementia, Parkinson’s disease, schizophrenia, anxiety, and depression) place a heavy societal and financial strain on the world’s healthcare systems. Recent research indicates that exposure to air pollution may hasten the onset of neurodegenerative disorders like Alzheimer’s disease (AD) and Parkinson’s disease (PD). Mental disorders are characterized by an elevated risk of metabolic syndrome (dyslipidemia, abdominal obesity, hypertension, and hyperglycemia). However, this discovery has not been verified in neurodegenerative diseases. Hence, we are curious about whether unfavorable environmental factors can magnify the effects of specific genetic, metabolic factors on the emergence of neuropsychiatric disorders.
Project duration: The project is planned to take 3 years after data download.
Public health impact: We hope that the results of this study will help us to provide a more comprehensive explanation of the underlying mechanisms of neuropsychiatric diseases. Then, based on the findings of this study, we will establish more accurate prediction models, develop scientific guidelines, better evaluate and intervene in risk factors of neuropsychiatric diseases, and lessen the burden of disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-genetic-polymorphisms-with-thrombocytosis

Association of genetic polymorphisms with thrombocytosis

Last updated:
ID:
128996
Start date:
4 December 2024
Project status:
Current
Principal investigator:
Dr Ing Soo Tiong
Lead institution:
Peter MacCallum Cancer Centre, Australia

Our research project aims to uncover the genetic factors that contribute to a common blood condition called thrombocytosis. Thrombocytosis happens when there are too many platelets in the blood, potentially increasing the risk of dangerous blood clots. Although we know some of the causes, there are still many cases where we can’t pinpoint why it happens. The duration of this project is approximately 12 months. By understanding the genetic cause of thrombocytosis and its association with blood clots, the doctors will be able to more accurately diagnosis and predict the risk of blood clots such as heart attack and stroke, and to implement appropriate treatment.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-genetic-variation-in-pltp-phospholipid-transfer-protein-with-mortality-from-sepsis

Association of genetic variation in PLTP (Phospholipid Transfer Protein) with mortality from sepsis.

Last updated:
ID:
77755
Start date:
8 February 2024
Project status:
Current
Principal investigator:
Dr Thomas Gautier
Lead institution:
Université de Bourgogne, France

Bacteria might induce severe infections, especially through the production of toxins that exert noxious effects upon human health by triggering inflammation. These toxins can be eliminated by the human organism, especially through circulating lipoproteins, the particles in charge of cholesterol transport in the blood. We have previously demonstrated that the phospholipid transfer protein (PLTP) enabled to enhance bacterial toxin removal and was protective upon sepsis induced mortality in animal models. We have demonstrated that this protein was associated with bacterial toxin concentration during cardiac surgery in humans. In the present study, we aim to explore whethehr genetic modifications of the gene that code for this protein are associated with the occurence of sepsis and sepsis induced mortality in order to further validate our hypothesis.
This work is an essential step toward the identification of new markers and the developement of therapeutics centered on this detoxification pathway to treat sepsis.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-genetic-variation-in-the-natriuretic-peptide-system-with-metabolic-risk-factors-cardiovascular-events-and-death

Association of Genetic Variation in the Natriuretic Peptide System with Metabolic Risk Factors, Cardiovascular Events and Death

Last updated:
ID:
58838
Start date:
17 July 2020
Project status:
Current
Principal investigator:
Dr Pankaj Arora
Lead institution:
University of Alabama at Birmingham, United States of America

Heart plays an endocrine role by secreting hormones called natriuretic peptides (NPs). These hormones regulate the cardiovascular physiology by controlling the blood pressure and how the body handles salt. These hormones also play a role in managing how the body processes glucose and generates energy. We and others have previously shown that common genetic variants of the NP genes result in changes in the circulating NP levels and resultantly in the blood pressure and cardiovascular physiology.

Variations in the circulating NP levels have been associated with development of adverse cardiovascular clinical events such as heart attacks, stroke, heart failure, arrhythmias, death due to heart disease and death due to any cause. We aim to examine the relationship of the various genetic variants that determine the circulating NP hormone levels, with the development of poor cardiovascular and metabolic health.

The project will span approximately 3 years and look at the various genetic variations which determine the changes in NP levels and their resultant impact on the cardiovascular health. The project will also involve the development of risk score, so that one can determine the likelihood of development of clinical disease and intervene pre-emptively to prevent or delay the development of the disease.

The proposed research will help improve the general understanding of the NP system and its close interplay in regulating the cardiovascular physiology and development of cardiovascular diseases. The proposed research will help in development of novel therapeutic strategies targeting the NP system. This will help in preventing the development of cardiovascular and metabolic diseases in individuals who are at an increased risk of such disease due to their genetic characteristics.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-genetic-variations-and-respiratory-disease-with-outcomes

Association of genetic variations and respiratory disease with outcomes

Last updated:
ID:
276802
Start date:
19 December 2024
Project status:
Current
Principal investigator:
Dr Vickram Tejwani
Lead institution:
Cleveland Clinic Foundation, United States of America

Chronic respiratory diseases are a major cause of morbidity and mortality, particularly chronic obstructive pulmonary disease (COPD) which is the third leading cause of death worldwide and asthma. These chronic respiratory diseases have varying severity of presentations and are also associated with other chronic diseases such as malignancies and cardiac disease. We propose studying genotypic and other demographic contributions to disease development among individuals with chronic respiratory disease to identify biologic correlates of disease severity and risk for other chronic diseases. This will serve to identify those at risk and based on the biologic pathways implicated, identify potential treatment targets for highly prevalent disease phenotypes. Thus, this project is in line with UK Biobank’s purpose to aid in the prevention, diagnosis, and treatment of illnesses and to promote health.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-genetics-biomarkers-and-environment-with-cognitive-function-and-mental-health

Association of genetics, biomarkers, and environment with cognitive function and mental health

Last updated:
ID:
23482
Start date:
1 November 2016
Project status:
Closed
Principal investigator:
Professor Helgi Schiöth
Lead institution:
Uppsala University, Sweden

This project primarily aims to investigate the interactions between genetics and environmental factors in relation to mental health and cognitive decline. We will study genetic variants with known associations to cognitive decline as well perform a genome-wide exploratory approach for novel variants to determine genetic contributions to mental health and its interaction with environmental variations. Furthermore, we would like to examine associations between known and novel biomarkers, and cognitive function to understand the biological pathways underlying susceptibility to impaired cognitive function. The project provides insights in the relative contribution that environmental factors have on cognitive functioning in adult individuals. This could contribute to public health policies to support brain health and cognitive function in later life. Furthermore, novel insights into the molecular biology and the genetic and epigenetic mechanisms underlying these associations will be critical for providing information to health professionals to ease public-health related decisions. Statistical models will be utilized to analyze the associations between genotype and cognitive function and mental health. To examine causal effect of these genes, Mendelian randomization will be applied. Novel genetic variants will be derived from two-thirds of the cohort and validated in the remaining third. Linear models will be used to study the association between environmental factors and cognitive function. Modifying effects of biomarkers on cognitive function will be assessed by incorporating it through generalized linear models. To maximize power, the full cohort will be included in the project.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-gluten-intake-with-metabolic-syndrome-and-cardiovascular-risk-findings-from-the-uk-biobank

Association of gluten intake with metabolic syndrome and cardiovascular risk – Findings from the UK biobank

Last updated:
ID:
47144
Start date:
20 February 2019
Project status:
Closed
Principal investigator:
Professor Mathias Fasshauer
Lead institution:
Justus Liebig University of Giessen, Germany

Although study results are contradictory, gluten intake has been linked to increasing health risks. Therefore, gluten-limited and gluten-free diets gained popularity also among healthy people without coeliac disease. The main reason for buying gluten-free products is that they are considered to be healthier than their conventional counterparts. However, a gluten-free diet may result in limited food choice and an unbalanced diet which is low in iron, zinc, folate, niacin, and fibre. Furthermore, to the best of our knowledge dietary gluten intake of UK Biobank participants has not been assessed so far.

This research aims to assess:
– The average dietary gluten intake of UK Biobank participants (aim 1)
– Whether gluten intake is associated with facets of the metabolic syndrome and vascular disease (cross-sectional study; aim 2)
– Whether gluten intake predicts all-cause and cardiovascular mortality (prospective study; aim 3)

In summary, the study will provide further evidence whether or not a gluten-limited or gluten-free diet is part of a healthy lifestyle. The research project is in line with the aims of UK biobank, i.e. to improve the prevention of serious public health risks like metabolic syndrome and cardiovascular disease.

We expect a project duration of 12 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-immune-gwas-loci-with-brain-structure-function-and-mental-health

Association of immune GWAS loci with brain structure, function, and mental health

Last updated:
ID:
48096
Start date:
29 May 2019
Project status:
Closed
Principal investigator:
Dr Michael Gandal
Lead institution:
University of California, Los Angeles, United States of America

Immune system dysregulation has been observed across several neuropsychiatric disorders including depression, bipolar disorder, and autism; however, whether or how immune alterations relate to brain-based changes associated with mental illness is unknown. One mechanism by which immune function may affect brain structure is through elimination of connections between neurons, called synapses. Indeed, altered brain volume and connectivity have been observed using magnetic resonance imaging (MRI) in many psychiatric disorders. Using data collected by the UK Biobank, this study seeks to investigate the relationship between neuroimaging measures of brain structure and connectivity, genetic predisposition to immune dysfunction, and mental health.

Our study has two aims. First, we will assess the additive impact of genetic risk for immune dysfunction across all genes – referred to as a polygenic risk score (PRS) – on MRI measures of brain volume, connectivity, and mental health. Furthermore, we will examine how exposure to stress may enhance the effects of immune-related PRS on the brain, leading to enhanced brain alterations in a subgroup of people with high PRS. Second, we will assess how changes in mental health across time might be predicted by measures of brain structure, PRS, and stress at baseline.

The results of this project may help to identify if certain individuals may be at higher risk for developing psychiatric illness, and eventually contribute to methods for the prediction of people in need of early mental health intervention.

We expect the duration of this project to be 36 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-immune-mediated-diseases-with-cancer-and-skin-diseases

Association of immune-mediated diseases with cancer and skin diseases

Last updated:
ID:
129011
Start date:
25 July 2025
Project status:
Current
Principal investigator:
Dr Minxue Shen
Lead institution:
Central South University, China

Immune-mediated diseases constitute a clinically heterogeneous group of disorders which jointly affect 5-10% of the population in developed countries. Several immune mechanisms for immune mediated diseases are also involved in cancer and skin diseases, such as inflammation-promoting TH17 dominance, dysfunctional Treg surveillance, and microbiota cross talk between these diseases. Many cancers and skin diseases, for reasons that are not known, are increased in patients with immune-mediated diseases patients. Moreover, decreased or increased survival was observed in patients with both immune-mediated diseases and cancer. However, small sample size and conflict results highlight the significance of a population-based prospective studies to clarify the association of immune-mediated diseases with cancer and skin diseases in incidence and survival. This proposed study aims to investigate the association of immune-mediated diseases with cancer and skin diseases in incidence and survival. We expect to find that immune-mediated diseases are both associated with risk and survival in cancer. We also expect that immune-mediated diseases are both associated with risk and survival in skin diseases. We also expect that skin disease are associated with risk and survival in cancer and immune-mediated diseases. The estimated study duration of our project is 36 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-impaired-vision-with-systemic-metabolic-endocrine-related-factors-and-lifestyle

Association of Impaired Vision with Systemic Metabolic/Endocrine-Related Factors and Lifestyle

Last updated:
ID:
107451
Start date:
31 August 2023
Project status:
Current
Principal investigator:
Professor Kun Liu
Lead institution:
Shanghai General Hospital, China

Aims: We aim to explore the correlation of impaired vision with systemic metabolism and lifestyle, and to investigate possible mechanisms of metabolic and ocular diseases.
Scientific rationale: Impaired vision is caused by a variety of ocular and systemic diseases, which have a serious impact on the quality of life. Congenital factors or lifestyle habits may lead to metabolic disorders, resulting in various endocrine disorders such as diabetes, hyperthyroidism and hyperlipidaemia. These diseases can cause damage to multiple tissues and organs, including the eyes, endocrine system and urinary system. In addition, ocular complications can contribute to different degrees of visual impairment, from refractive changes to complete blindness. Despite treatment, some patients may still experience the development or exacerbation of ocular complications at different stages of the disease. The hypothesis posits a robust correlation between ocular health and systemic metabolism. Numerous studies in metabolomics, proteomics, and lipidomics have substantiated that ocular diseases undergo metabolic alterations not only in the eye but also in the blood and urine. We aim to investigate whether systemic metabolic/endocrine-related indicators and lifestyle are associated with impaired vision and how risk factors interact with impaired vision. Moreover, potential mechanisms will be explored through multivariate linear/logistic regression, mediation analysis and Mendelian randomisation methods.
Project duration: 36 months.
Public health impact: Our project will benefit hundreds of millions of people worldwide who are suffering from visual impairment. In the event that metabolic and lifestyle-related risk factors are discerned, lifestyle modifications may present themselves as promising targets for intervention, thereby engendering novel approaches to the prevention and management of systemic diseases that give rise to ocular complications. Additionally, the examination of the correlation between metabolomics, genomics, imaging, lifestyle, and ocular diseases may furnish fresh insights into the identification of potential therapeutic targets and follow-up indicators.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-intra-ocular-pressure-iop-with-genetic-variation-in-the-genes-hcrt-or-hcrtr1-or-hcrtr2

Association of intra-ocular pressure (IOP) with genetic variation in the genes HCRT or HCRTR1 or HCRTR2.

Last updated:
ID:
102073
Start date:
26 November 2024
Project status:
Current
Principal investigator:
Dr Arthur DeCarlo
Lead institution:
University of Alabama at Birmingham, United States of America

Glaucoma is one of the leading causes of blindness worldwide. While we know that increased eye pressure is a risk factor for glaucoma, recent evidence also indicates differences between eye pressure and the pressure in the brain may also be a risk factor. The research described in this application is aimed at investigating how centers within the brain control eye pressure and will support the goal of identifying new treatment options for patients with glaucoma. The orexin neurotransmitter system, with one prepro-peptide generating two neurotransmitter forms, and two separate orexin receptor genes, may have a role in intraocular pressure (IOP) regulation and we aim to study variability in those genes. Orexin receptor antagonists are taken worldwide for insomnia and the discovery proposed here could warrant addition of a new data field reporting prescribed use of orexin receptor antagonists. The analysis of this dataset is expected to require 2 years working with co-investigators in genomics and statistical analysis.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-lifestyle-behaviors-and-genetic-risk-with-cardio-cerebrovascular-diseases

Association of Lifestyle Behaviors and Genetic Risk With cardio-cerebrovascular diseases.

Last updated:
ID:
90311
Start date:
14 September 2022
Project status:
Current
Principal investigator:
Professor Minghuan Wang
Lead institution:
Tongji Hospital, China

Cardio-cerebrovascular disease is the major cause of mortality and morbidity and is driven by both genetic and environmental factors. Early evidence supporting a role for genetics in risk of a range of vascular events came from genome wide association studies. Lifestyle is an another important modifiable risk factor for vascular events. Clear evidence has showed that adhering to a healthy lifestyle, including not smoking, regular physical activity, and a healthy diet, is associated with a decreased risk of stroke and cardiovascular disease.
The results indicated that genetic and lifestyle factors were independently associated with risk of cardio-cerebrovascular diseases, and an unfavorable lifestyle profile was associated with increased risk of cardiovascular disease and stroke across all genetic risk stratums. Further evidence indicated that genetic composition and combined health behaviors had a log-additive effect on the risk of developing cardiovascular disease and stroke and adhering to a healthy lifestyle could attenuate the effect of genetics on cardiovascular disease and stroke risk. However, the relationship with genetic and lifestyle risks in cardio-cerebrovascular diseases have not yet been fully investigated, and the extent to which this can be offset by lifestyle factors is unknown.
In this study, we plan to conduct a Mendelian randomization (MR) analysis to assess the causal effects of lifestyle behaviors on risk of cardio-cerebrovascular disease and investigate whether health behaviors may offset the genetic risk for age-related health outcomes. We hope our study will provide a unique evidence regarding the causal relationships between lifestyle behaviors and cardio-cerebrovascular diseases, which may help to identify those at high risk of vascular events at an early stage and highlight the potential of lifestyle interventions to reduce risk of vascular events across entire populations, even in those at high genetic risk.
The project is scheduled to begin in July 2022 and be completed in December 2023. The research plan are as follows:
1) July 2022-December 2022: Genome-wide association analysis identifies genetic variations associated with lifestyle behaviors and cardio-cerebrovascular disease.
2) January 2023-June 2023: (1) To conduct a Mendelian randomization study to assess the causal effects of lifestyle behaviors on risk of cardio-cerebrovascular disease. (2) To investigate whether a healthy lifestyle is associated with lower risk of cardio-cerebrovascular disease regardless of genetic risk.
3) July 2023-December 2023: Summarizing research results, writing research papers.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-lifestyle-behaviors-with-incident-and-all-cause-mortality-of-cardiovascular-kidney-metabolic-syndrome

Association of lifestyle behaviors with incident and all-cause mortality of cardiovascular-kidney-metabolic syndrome

Last updated:
ID:
590955
Start date:
25 June 2025
Project status:
Current
Principal investigator:
Professor Xiaoyong Yu
Lead institution:
Shaanxi Provincial Hospital of Traditional Chinese Medicine, China

Obesity, diabetes, and chronic kidney disease (CKD) are each associated with a high burden of cardiovascular disease (CVD) morbidity and mortality; they commonly co-occur and disproportionately affect disenfranchised populations. Recently, the American Heart Association (AHA) has characterized cardiovascular-kidney-metabolic syndrome (CKM Syndrome) as the interactions and pathophysiological connections among the cardiovascular system, renal system, and metabolic risk factors, leading to multiorgan dysfunction and to a complex clinical presentation. In the USA, more than 1 in 4 adults present at least one condition of the triad, while the prevalence of concurrent comorbidities associated with CKM is approximately 25 -30% worldwide, reflecting the seriousness of this multimorbidity condition.The clinical implications of poor CKM health are significant, with potential for mortality, excess morbidity, multiorgan disease, and high health care expenditures driven largely by the burden of cardiovascular disease (CVD).The study recently showed that cumulative social disadvantage, denoted by higher social risk profile (SRP) burden, was associated with higher odds of CKM multimorbidity, independent of demographic and lifestyle factors. However, few studies have studied the association between lifestyle behaviors and CKM. Therefore, our current study aimed to investigate the LE8 score and incident and all-cause mortality of CKM syndrome from the UK Biobank.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-lipoprotein-profile-with-cardiovascular-disease-and-its-risk-factors

Association of lipoprotein profile with cardiovascular disease and its risk factors

Last updated:
ID:
100430
Start date:
19 April 2023
Project status:
Current
Principal investigator:
Mr Bradley James Tucker
Lead institution:
University of Sydney, Australia

Cardiovascular disease (CVD) is the leading cause of death globally. The previous two decades of cardiovascular research has largely focused on reducing ‘bad cholesterol’, also known as low-density lipoprotein cholesterol. Several therapeutic interventions have been developed to effectively reduce this bad cholesterol, however, even in those patients with normal or low levels of bad cholesterol the incidence of cardiovascular disease remains unacceptably high. This phenomenon is termed residual risk and to further understand this residual risk we aim to use more detailed lipoprotein measures. Nuclear magnetic resonance (NMR) spectroscopy is able to provide a comprehensive analysis of not only total plasma cholesterol or triglyceride concentration but also precise details on lipoprotein particle size, number of particles and their composition. In smaller cohorts, NMR-derived lipoprotein parameters have been shown to have superior predictive power compared to standard lipid measurements for future CVD events. However, as of yet no one has confirmed these findings in a large, diverse cohort such as that of the UK Biobank.

Therefore, the aim of this study is to comprehensively assess the relationship of lipoprotein distribution with CVD and its risk factors. Risk factors for CVD that will be investigated in this study included diabetes, inflammation and subclinical atherosclerosis. This project should take approximately 24 months to complete.

By providing robust evidence on the association of lipoprotein distribution with CVD and its risk factors, results of this study will help better understand the mechanisms of disease at play in CVD. Better understanding of disease mechanisms will aid drug development and hopefully enhance therapeutic options available for the treatment/prevention of CVD. Moreover, by testing the predictive power of various lipoprotein variables we aim to improve risk prediction for CVD in both the primary and secondary prevention settings. This will facilitate early intervention of disease and ultimately improve long-term health outcomes for many.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-loneliness-and-social-isolation-with-postoperative-outcomes

Association of Loneliness and Social Isolation with Postoperative Outcomes

Last updated:
ID:
116151
Start date:
5 October 2023
Project status:
Current
Principal investigator:
Ms Kaylyssa Philip
Lead institution:
University of Toronto, Canada

Over 40% of older adults regularly experience loneliness. Prior research has shown that loneliness and social isolation is associated with a greater risk of medical conditions such as cardiovascular disease, dementia and premature death. Furthermore, low functional mobility and depression is associated with increased postoperative mortality. However, whether loneliness impacts postoperative outcomes is uncertain. It is known that patients undergoing nonelective surgery are particularly at risk of loneliness. Therefore, identifying the effect of loneliness and social isolation in surgical patients is important to optimize perioperative support.

This research project will study whether loneliness and social isolation impact surgical outcomes, particularly 90-day mortality and 30-day postoperative complications. The expected project duration is one year.

The proposed research project will provide valuable information to the public. Specifically, it will help determine if loneliness and social isolation adversely impact postoperative outcomes, which represents an important gap in the literature. Given the high prevalence of loneliness, it is crucial that the consequences are understood and quantified. Depending on the results, the findings may also provide evidence for enhanced perioperative interventions among these patients, such as closer postoperative follow-up and peer support models.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-loss-of-function-variants-and-incompletely-penetrant-alleles-with-ukbb-phenotypes

Association of loss-of-function variants and incompletely penetrant alleles with UKBB phenotypes

Last updated:
ID:
42890
Start date:
22 February 2019
Project status:
Closed
Principal investigator:
Daniel MacArthur
Lead institution:
Broad Institute, United States of America

Even though we have created genetic data for a very large number of people, we still don’t know the clinical effects of most of the genetic differences between people. We propose to investigate the outcomes of two important categories of genetic differences:
Loss of function variants – genetic differences predicted to cause a gene to partially or completely stop working; and
Incompletely penetrant variants – genetic differences that cause severe diseases in some people, but not in others
We propose to spend the next 3 years investigating those two categories of genetic differences in the participants in the UK BioBank. From this, we hope to learn about how possessing a broken copy of a gene affects people and how to predict the odds of getting severe diseases from a person’s genetic data. We hope that this will help scientists who want to understand what the functions of genes are, drug developers who want to predict the side effects of the drugs they are developing, and doctors who want to advise people with genetic data on what their odds are of developing diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-male-infertility-genotypes-on-mortality-cancer-and-cardiovascular-disease

Association of male infertility genotypes on mortality, cancer, and cardiovascular disease

Last updated:
ID:
53354
Start date:
2 October 2019
Project status:
Current
Principal investigator:
Dr Michael Eisenberg
Lead institution:
Stanford University, United States of America

Infertile men have been shown to be sicker than fertile men. They have higher risk of early death and a variety of severe diseases. There are many forms of infertility and some have been associated with certain genes and mutations. It is, however, unknown whether the infertility-related genes also lead to these increased risks of disease, or if infertility is related to health through other mechanisms (e.g. lifestyle, diet, etc.). Therefore, the aim of this study is to investigate the risk of mortality and the development of comorbidities for men possessing DNA signatures consistent with male infertility compared to those without those genotypes within the UK Biobank population. By doing so, we will be able to deduce whether some genotypes predispose to male infertility and future disease and how large of contribution towards higher risk they incur. This information can be useful as a first step towards understanding why these men have higher disease risk and potentially allow better patient counseling.
The methodology that we propose will start by identifying which DNA signatures are associated with male infertility. We will then link these DNA signatures to the UK biobank data to determine the association with later health problems.
The total project duration is estimated to take 24 months. This includes 6-9 months of analysis, 3-5 months of article writing, and 3-10 months of article review.
We consider this project to potentially have a large positive public health impact. Men with infertility constitute a large proportion of all men and this group has historically received little attention even with the large health risks they face. Other studies, mostly epidemiological studies, have already established these high risks of disease for infertile men, ranging from higher risk of cancer to earlier death. The proposed study will focus on understanding the mechanisms involved and especially the contribution of genetics toward these risks. This has not been studied thus far, and therefore could substantially improve the understanding of these high risks. If a genetic link is established, this will allow better patient screening and counseling as infertile men present for care.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-malignant-tumor-with-toxoplasma-gondii-infection

Association of malignant tumor with Toxoplasma gondii infection

Last updated:
ID:
71509
Start date:
8 September 2021
Project status:
Current
Principal investigator:
Miss Min Chen
Lead institution:
Southern Medical University, China

Toxoplasma gondii (T. gondii) infection is becoming a major cause of morbidity and mortality in immunocompromised patients such as malignant tumor patients. However, the disease burden of T. gondii infection in the malignant tumor patients is poorly understood. Therefore, we will use data obtained from the UK Biobank to evaluate the incidence, treatment, prognosis of malignant tumor patients co-infected with T. gondii, and to predict risk factors associated with the prevalence of T. gondii infection in these populations. We aim to complete this project within one year. The results of this research will contribute to the future scientific researches in this area, and help us understand the situation of T. gondii infection in malignant tumor patients; so as to emphasize the importance of routine surveillance of T. gondii infection in these populations, as well as the information targeting preventive measures and treatment strategies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-melanoma-and-non-melanoma-skin-cancers-with-the-interaction-between-lifestyle-and-genetic-factors

Association of melanoma and non-melanoma skin cancers with the interaction between lifestyle and genetic factors

Last updated:
ID:
55257
Start date:
12 June 2020
Project status:
Current
Principal investigator:
Dr Hong Liu
Lead institution:
Central South University, China

We aim to investigate the impact of lifestyle, genetic factors and their interaction with the incidence of skin cancers. Previous literature has suggested that physical activity is protective against various cancer types in both smokers and non-smokers. However, systemic exploration of how the beneficial influence of physical activity might be attenuated or strengthened by genetic background is limited. With the large sample size of UK Biobank, we will have sufficient power to characterize this potential interaction. In addition, blood biochemistry markers have been a proxy for organ function and widely used in the clinic. We would like to investigate whether these markers can be used to inform the interaction between lifestyle factors and genetic composition.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-menstrual-and-reproductive-factors-with-the-risks-of-cancer

Association of menstrual and reproductive factors with the risks of cancer

Last updated:
ID:
141156
Start date:
27 October 2023
Project status:
Current
Principal investigator:
Professor Shipeng Yan
Lead institution:
Hunan Cancer Hospital, China

In this project, we will identify the associations of menstrual factors and reproductive history with the risks of cancer and its subtypes. Menstrual factors included age at menarche, age at menopause, length of menstrual cycle, using of hormone-replacement therapy, use of oral contraceptive pill, etc. Reproductive factors included age at first live birth, history of stillbirth, spontaneous miscarriage or termination, number of stillbirths, etc. Sex hormones, such as estrogen and progesterone, are important agents in female cancer development and progression. Given this premise, female menstrual and reproductive factors, as remarkable indicators of hormone effect, were hypothesized to be associated with cancer risk. However, the existed epidemiological evidence was inconsistent or limited to certain female-specific cancer subtypes. The UK Biobank, a large population-based prospective study, may provide more comprehensive and reliable evidence in terms of the above associations. In addition, we can use hormone data from UK Biobank to further investigate the role of hormone levels on the association between menstrual and reproductive factors and cancer, particularly endocrine cancers. The project is planning to take 3 years after the data has been downloaded. Understanding the link between menstrual and reproductive factors and cancer risk in women can have implications for public health policies, prevention strategies, and individualized healthcare decisions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-mental-health-on-multimorbidity-psychiatric-medications-behavioral-factors-and-multiple-disease-outcomes

Association of mental health on multimorbidity: psychiatric medications, behavioral factors and multiple disease outcomes

Last updated:
ID:
106402
Start date:
31 August 2023
Project status:
Current
Principal investigator:
Dr Wai Kai Hou
Lead institution:
The Education University of Hong Kong, Hong Kong

This 36-month research project aims to explore how mental health affects the development of physical health in individuals with multimorbidity. The mechanisms between mental health, psychiatric medications, behavioral factors, and multiple physical diseases will be studied. Previous studies suggested that mental health potentially affects the development and progression of physical health outcomes. However, little is known about the mechanisms behind the association. Therefore, this study will explore (1) the impact of psychiatric medications on physical disease outcomes, and (2) how behavioral factors like lifestyle, stress resilience, and spousal interactions would influence physical health.

The research findings have the potential to highlight the significant role of mental health on physical health outcomes. By understanding the mechanism between mental health and physical health, we aim to improve the early recognition and management of mental health issues in individuals with multimorbidity. This research may also contribute to an increased awareness of mental well-being by optimizing psychiatric medication use and improving stress resilience patterns. Ultimately, the goal is to improve the well-being of individuals with multimorbidity.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-metabolic-factors-and-medication-use-with-the-risk-and-progression-of-knee-osteoarthritis

Association of Metabolic Factors and Medication Use with the Risk and Progression of Knee Osteoarthritis

Last updated:
ID:
649025
Start date:
26 May 2025
Project status:
Current
Principal investigator:
Dr Xianjie Wei
Lead institution:
Beijing Tsinghua Changgung Hospital, China

Research Questions:
How do metabolic factors such as obesity, hyperglycemia, and dyslipidemia contribute to the onset and progression of knee osteoarthritis (KOA)?
What is the role of medications for metabolic conditions (e.g., statins, glucose-lowering agents) in modifying KOA risk and progression?
Can predictive models incorporating metabolic and medication-related data accurately identify individuals at high risk of KOA and guide personalized prevention and treatment strategies?
Objectives:
To evaluate the associations between key metabolic factors and the risk of KOA.
To investigate the impact of metabolic-related medications on KOA progression and outcomes.
To develop and validate predictive risk models using metabolic and pharmacological data for personalized KOA management.
Scientific Rationale:
Knee osteoarthritis (KOA) is a leading cause of disability globally, with metabolic syndrome identified as a significant contributor to its pathogenesis through mechanisms like inflammation and oxidative stress. However, the interaction between metabolic factors and medications in modifying KOA outcomes remains underexplored. By leveraging the extensive data from the UK Biobank, this study aims to uncover these relationships, identify biomarkers, and advance precision medicine for KOA management.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-metabolomic-profiles-with-mri-traits-of-cardiac-structure-and-function-and-with-the-risk-of-incident-heart-failure-in-uk-biobank-cohort

Association of Metabolomic profiles with MRI traits of cardiac structure and function and with the risk of incident heart failure in UK Biobank Cohort

Last updated:
ID:
75001
Start date:
23 September 2021
Project status:
Closed
Principal investigator:
Mr Jakub Morze
Lead institution:
University of Warmia and Mazury in Olsztyn, Poland

Aims: Our study aims to use data from the UK Biobank cohort to evaluate the role of metabolism disruption in heart failure (HF). To this end, we will look at the relationship between metabolite levels and 1) abnormalities of heart function and structure, 2) risk of developing HF. Additionally, we will explore potential differences in the metabolism between individuals with HF who simultaneously have other underlying diseases, such as coronary heart disease, cardiomyopathy, and arrhythmia. These disorders increase the likelihood of developing HF.
Scientific rationale: HF is a syndrome in which the heart is not able to provide enough output to meet the body’s demand for oxygen. It is a chronic complication of various cardiovascular diseases. Improved cardiovascular care contributed to reduced mortality of cardiac patients and increased the number of individuals at risk of HF onset. Deteriorating heart function reduces exercise capability and tolerability, leading to a substantial decrease in quality of life. Only about half of the patients diagnosed with HF survive five years after the diagnosis has been made, indicating generally poor survival for patients with HF. Previous research indicated that measurement of particular biochemical markers could predict heart dysfunction many years before onset.
However, currently clinically used biomarkers of HF (such as BNP) rather reflect physiological adaption of the heart during the disease process rather than early changes leading to the development of disease. Therefore, there is a substantial need to identify early biochemical indicators of the different disease processes leading to HF onset. The metabolome represents the totality of compounds involved in human metabolism. By comparing the baseline metabolome of patients who developed HF in longitudinal observation compared to those who remained healthy during the observation period, we can suggest potential metabolic indicators of disease. Findings from previous research on metabolome in HF are not conclusive and limited by a small samples size or by study design.
Duration: This project will last 36 months upon receipt of the data.
Public health impact: Despite novel treatment opportunities, HF still remains a crucial public health issue with approximately 64 million affected individuals worldwide. Our study will provide insight into metabolic pathways reflecting processes involved in the onset of HF. Our planned analyze might contribute to identifying potential markers of individuals at high risk of HF onset, enabling effective prevention. This can be a step toward personalized care of HF, addressing individual characteristics and needs of patients.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-migraine-and-cardiac-arrhythmias

Association of migraine and cardiac arrhythmias

Last updated:
ID:
394593
Start date:
9 January 2025
Project status:
Current
Principal investigator:
Dr Le Li
Lead institution:
Chinese Academy of Medical Sciences &Peking Union Medical College, China

Migraine is a severe headache disorder that significantly affects quality of life, particularly in women under 50 years old. It is one of the leading causes of disability due to the high number of years lived with disability. Previous studies have shown that migraines, especially those with aura, are associated with an increased risk of cardiovascular diseases. However, the relationship between migraines and cardiac arrhythmias, such as atrial fibrillation, bradyarrhythmias, and ventricular arrhythmias, is not well understood.

Our study aims to investigate whether individuals with migraines are at a higher risk of developing cardiac arrhythmias compared to those without migraines. We will also examine if genetic susceptibility and commonly used migraine medications influence this risk. Participants will be classified as having migraine based on a clinical diagnosis or self-reporting of migraine. Those with pre-existing cardiovascular diseases at baseline or insufficient data on key covariates will be excluded from the study.

To analyze the data, we will calculate the cumulative incidence of cardiac arrhythmias using the Kaplan-Meier method and compare differences between individuals with and without migraines using the log-rank test. Cox proportional hazards regression models will be used to assess the association between migraines and the risk of cardiac arrhythmias. Sensitivity analyses, including excluding participants with relevant comorbidities and using propensity score matching, will be conducted to ensure the robustness of our findings.

Additionally, we will calculate polygenic risk scores for cardiac arrhythmias to evaluate the role of genetic susceptibility in the migraine-arrhythmia relationship. We will also perform a summary data-based Mendelian randomization analysis to explore the effect of migraine medications on the risk of cardiac arrhythmias.

The project will last for approximately three years. The results of this research could lead to improved screening and management strategies for cardiac arrhythmias in individuals with migraines, ultimately reducing morbidity and healthcare costs, and improving public health outcomes.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-multiple-environmental-exposures-and-non-environmental-factors-with-cancer-incidence-and-mortality

Association of multiple environmental exposures and non-environmental factors with cancer incidence and mortality

Last updated:
ID:
226023
Start date:
25 July 2025
Project status:
Current
Principal investigator:
Ms Yan Zhao
Lead institution:
The First Hospital of China Medical University, China

Cancer is emerging as a major public health challenge globally and also a complex and multifactorial disease. The acute impact of climate change on human health is receiving increased attention, but little is known or appreciated about the effect of climate change on chronic diseases, particularly cancer. Epidemiological studies over the past two decades have provided strong evidence that environmental components interacting with lifestyle factors can individually and collectively influence the occurrence of cancer. However, prior research has largely focused on studying exposures to one contaminant at one time, which does not reflect the real-world environment. In addition, few studies have investigated the combined and synergistic effects of environmental exposure and non-environmental factors on incidence and mortality of different cancers. The present study aimed to assess the associations of multiple environmental exposures (e.g. residential air pollution, residential noise pollution, and greenspace) and non-environmental factors (e.g. modifiable lifestyle factors, biochemical markers, medication use, early life factors, and comorbidities) with incidence and mortality of different cancers (e.g. breast cancer, ovarian cancer, cervical cancer, gastric cancer, and liver cancer) and to investigated the combined and synergistic effects of environmental exposure and non-environmental factors on incidence and mortality of different cancers. This project has important implications for improving health status and developing strategies to prevent cancers.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-multiple-environmental-exposures-with-adverse-health-impacts-and-their-interaction-with-genetics-and-diet

Association of multiple environmental exposures with adverse health impacts and their interaction with genetics and diet

Last updated:
ID:
97753
Start date:
30 November 2023
Project status:
Current
Principal investigator:
Professor Yunfeng Zou
Lead institution:
Guangxi Medical University, China

It is well-known that exposure to a polluted environment (e.g., air pollution and noise pollution) poses a great health risk to humans. For example, approximately 4.51 million deaths worldwide in 2019 was attributed to exposure to ambient air pollution. Exposure to ambient air pollution has been associated with elevated morbidity and mortality of respiratory, cardiovascular, kidney, and neurological diseases. Although it is well established that expression of certain genes makes humans more susceptible to environmental exposure induced adverse health impacts, the underlying mechanisms involving such genetic susceptibility remain unclear. In addition, research has been taken to investigate potential interventional strategies at an individual level to mitigate adverse effects of environmental exposures. Dietary components, such as vegetable and fish intake, have been suggested to confer health benefits against exposure to air pollution. There is also emerging evidence showing health benefits of exposure to natural greenness. However, compelling evidence from a large perspective study is lacking to validate these findings.
In this proposal, we will use the ample data from the UK Biobank to investigate the genetic susceptibility and modifications of environmental exposures and adverse disease outcomes. Specifically, we aim to establish association between environmental exposures and health outcome parameters of selected diseases, investigate the gene-environment interactions on the disease outcome variables, and assess the modifying effects of dietary components or greenness on the established environment – health outcome associations. The successful completion of this 36- month proposal will likely contribute to our knowledge on the genetic mechanisms of environmental pollution induced health effects and mitigation strategies. We expect to identify several key genetic pathways that are essential in the environmental exposure induced health outcomes and verify health benefits of certain dietary components or greenness against environmental exposures. The findings of this proposal will provide key scientific evidence for the policy makers to consider intervention strategies to mitigate adverse health effects of environmental exposures at an individual level.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-neurodegenerative-diseases-with-cancer

Association of neurodegenerative diseases with cancer.

Last updated:
ID:
93435
Start date:
16 January 2023
Project status:
Current
Principal investigator:
Professor Yuan Shen
Lead institution:
Tongji University School of Medicine, China

Cancer and neurodegenerative diseases, such as Alzheimer’s disease, Parkinson’s disease and Huntington’s disease, are among the leading causes of human death around the world. Research has found a potential inverse association between cancer and neurodegenerative diseases. Specifically, patients with neurodegenerative diseases may have a lower risk of cancer and those with cancer may have a lower risk of neurodegenerative diseases. However, different findings exist and the previous studies could not study the association systematically due to data limitations. Moreover, the pathogenesis of both cancer and neurodegenerative diseases is largely unknown and less is known about the biological mechanism between the two diseases.

We, therefore, propose the research project to comprehensively investigate the association between neurodegenerative diseases and cancer. With the help of UK biobank database, the project will analyze the association between different types of neurodegenerative diseases and different types of cancers with appropriate statistical methods. The project will also investigate the effect of treatment (e.g., surgery, chemotherapy, drugs, etc.) for one disease on the risk of other disease. If the inverse association between neurodegenerative diseases and cancer could be verified, the further intention of this project is to identify the brain structure changes and genetic alterations in the inverse association between neurodegenerative diseases and cancer.

The estimated duration of this project is 36 months, but due to the complex nature of the analysis, it may need to be extended for a longer period.

This project could help in understanding the complicated relationship between neurodegenerative diseases and cancer, revealing innovative pathogenesis regarding neurodegenerative diseases and cancer, and developing a strategic plan for preventing or delaying cancer and/or neurodegenerative diseases onset.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-night-shifts-workgenetic-risk-and-risk-of-gastrointestinal-cancer-in-the-uk-biobank

Association of night shifts work!genetic risk and risk of gastrointestinal cancer in the UK Biobank

Last updated:
ID:
91734
Start date:
16 September 2022
Project status:
Current
Principal investigator:
Dr Xiawei Li
Lead institution:
Zhejiang University, China

1.Aims: We aim to examine the effects of past and current night shift work on gastrointestinal cancer and whether night shifts work modifies the genetic gastrointestinal cancer predisposition.

2. Scientific rationale: Approximately 15-20 % of the working population in industrialized countries is estimated to engage in night-shift work, including permanent night shifts, rotating shifts, and irregular schedules!and the effect of such a work schedule on health, including on the formation of cancers, has attracted increasing multidisciplinary research attention. Shift work, particularly night shifts, disrupts social and biological rhythms, as well as sleep, and through those pathways has been suggested to increase the risk of cancer.
In addition to environmental factors such as shift work, genetics also plays an important role in gastrointestinal cancer risk. According to some research, lifestyle and environmental factors may alter genetic susceptibility to chronic diseases. Thus, lifestyle and environmental factors may also alter genetic susceptibility to gastrointestinal cancers, a currently unexplored question.
3. Project duration: Three years will be needed to complete this project.
4. Public health impact: With the prevalence of gastrointestinal cancer on the rise, we need to understand which aspects of the shift work schedule are likely to be the most disruptive, and for which specific gastrointestinal cancers are positively correlated, which is critical for designing targeted primary and secondary prevention strategies. It ultimately could help reduce disease-related societal burden and economic cost. In addition, our study on whether night shift modifies genetic susceptibility to gastrointestinal cancer could genetically explain the association between night shift and gastrointestinal cancer, a question worth exploring.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-noise-pollution-and-cardiometabolic-disease

Association of Noise Pollution and Cardiometabolic Disease

Last updated:
ID:
69597
Start date:
25 January 2021
Project status:
Current
Principal investigator:
Dr Guang Hao
Lead institution:
Jinan University, China

Aims: To study the associations between noise pollution and cardiometabolic risk, and the roles of mental health and sleep quality in those associations.
Scientific rationale: The causes of cardiometabolic disease are complex and still not fully studied. Therefore, it is important to identify modifiable risk factors of cardiometabolic disease. Noise pollution is a common and under-recognized health risk. Traffic noise exposure is linked to cardiometabolic diseases such as type 2 diabetes, arterial hypertension, myocardial infarction, and stroke. However, the underlying mechanisms are not yet well understood.
Project duration: 24 month
Public health impact: Our research will clarify the associations between noise pollution and cardiometabolic disease, and the roles of mental health and sleep quality using this large cohort of UK biobank. The findings may help to build evidence-based strategies to reduce the burden of cardiometabolic disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-novel-heart-failure-risk-factors-and-biomarkers-with-incident-cancer

Association of novel heart failure risk factors and biomarkers with incident cancer

Last updated:
ID:
334205
Start date:
30 April 2025
Project status:
Current
Principal investigator:
Dr Yiqian Yang
Lead institution:
Erasmus MC, Netherlands

Rationale: Community-based prospective studies have demonstrated that HF risk factors, including age, sex, smoking status, and obesity, are also independent risk factors of cancer development. However, existing studies have mainly focused on the relationship between traditional HF risk factors and incident cancer, although emerging data clearly suggest that abundant novel risk factors may also lead to HF. Recently, our group conducted an analysis of a wide range of HF risk factors using UK Biobank data, and identified around 50 risk factors, both traditional and novel, associated with incident HF.

Aims: The main aim of this study is to examine the association between these newly identified HF risk factors and incident cancer.

Project duration: The expected project duration is 36 months.

Public health impact: HF and cancer pose a heavy burden on our healthcare system, and this burden will increase in the future owing to the aging of the population. Examining the association of novel HF risk factors and biomarkers with future incident cancer may lead to identify individuals at risk, inform the development of monitoring and interventional strategies to mitigate HF and cancer risks in the general population, and elucidate the potential biological pathways underlying both HF and cancer.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-nutrient-composition-with-metabolic-syndrome-and-cardiovascular-risk-findings-from-the-uk-biobank

Association of nutrient composition with metabolic syndrome and cardiovascular risk – Findings from the UK Biobank

Last updated:
ID:
53438
Start date:
5 November 2019
Project status:
Current
Principal investigator:
Professor Mathias Fasshauer
Lead institution:
Justus Liebig University of Giessen, Germany

Nutrition plays a key role in the prevention and treatment of obesity which is associated with metabolic disease including type 2 diabetes mellitus and cardiovascular disease. Extensive studies exist on total macronutrient intake in relation to obesity and metabolic health. However, there is a growing body of evidence that nutrient composition, i.e. relative distribution and components of nutrients consumed, rather than total macronutrient consumption affects metabolic and vascular health.

Therefore, the present project assesses whether nutrient composition is associated with:
– Features of the metabolic syndrome and vascular disease (cross-sectional study; aim 1).
– Incidence of specific health-related outcomes (prospective study; aim 2).
– All-cause and disease-specific mortality (prospective study; aim 3).

The present project will contribute to evidence-based recommendations on optimal dietary patterns and food composition to prevent metabolic and vascular disease. Therefore, the research project is in line with the aims of UK biobank, i.e. to improve the prevention of serious public health risks like metabolic syndrome and cardiovascular disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-objectively-measured-physical-activity-sedentary-behavior-and-sleep-patterns-with-degenerative-diseases

Association of objectively measured physical activity, sedentary behavior, and sleep patterns with degenerative diseases

Last updated:
ID:
778079
Start date:
23 May 2025
Project status:
Current
Principal investigator:
Ms Seounghee Choi
Lead institution:
Korea Maritime & Ocean University, Korea (South)

With the growing elder populations, the prevalence of age-related musculoskeletal and neurodegenerative diseases-including Parkinson’s disease and dementia-has been steadily increasing. These degenerative conditions pose significant therapeutic challenges due to their complex pathophysiological mechanisms and clinical heterogeneity. Given these therapeutic constraints, numerous studies emphasized early identification and prevention.
Previous research has suggested that lifestyle factors-such as physical activity, sedentary behavior and sleep-are associated with degenerative diseases and may play a crucial role in prevention. However, most of the studies have several limitations such as small sample size, short follow-up period and limited variables. For example, assessments of physical activity and sleep have often relied on self-reported questionnaires, which are susceptible to recall bias.
Moreover, few studies have comprehensively investigated the relationship between degenerative diseases and the objectively measured lifestyle variables including physical activity, sedentary behavior, and sleep. UKBiobank (UKB) has over 500,000 participants, and provides extensive data such as basic health related information, plasma, genetics and bioimaging as well as accelerometry data to assess lifestyle objectively.
Using UKB data, our primary aim is to examine the association between accelerometer-measured lifestyle factors-specifically physical activity, sedentary behavior, and sleep-and degenerative diseases. Our secondary aim is to predict disease progression by integrating multiple variables, including plasma biomarkers, genetic profiles, and brain structures linked to lifestyle patterns. This three-year project will commence upon application approval and will utilize the full range of UKB cohort data. Through the project, we aim to contribute to the development of lifestyle guidelines and prevention strategies for older adults at risk of degenerative diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-omega-3-fatty-acids-and-vital-substances-with-presence-and-rupture-of-human-intracranial-aneurysms

Association of omega-3-fatty acids and vital substances with presence and rupture of human intracranial aneurysms

Last updated:
ID:
411099
Start date:
8 November 2024
Project status:
Current
Principal investigator:
Dr Katharina Hackenberg
Lead institution:
Heidelberg University, Germany

Background:
Unruptured intracranial aneurysms (IAs) have a prevalence of 3% in the adult population and can lead to aneurysmal subarachnoid haemorrhage (SAH), which still carries a poor prognosis with high morbidity and case-fatality. Preventive treatment comes with a risk for stroke up to 4% and for death up to 0.3%. Since the risk of rupture in most incidental diagnosed IAs is less than the risk of preventive repair, conservative treatment options, that accompany observation with follow-up imaging, are urgently needed.
Recent studies showed that a high intake of omega-3-fatty acids lead to a reduction of stroke incidence, especially in women. On a molecular level omega-3-fatty acids exert different ways of anti-inflammatory effects, i.e. reduction in pro-inflammatory macrophages, recruitment of anti-inflammatory macrophages. Vital substances as Vitamin K2 seem to inhibit the differentiation of vascular smooth muscle cells (vSMCs) leading to reduced atherosclerosis and reduce pro-inflammatory mediators.
In pathogenesis of IAs inflammatory processes as recruitment of macrophages followed by degradation of the extracellular matrix as well as differentiation of vSMCs within the vascular wall play a major role leading to IA formation and IA rupture.

Aim:
The study aims to show the protective role of omega-3-fatty acids and vital substances on the pathogenesis of IAs. We hypothesize that the intake of omega-3-fatty acids and vital substances is associated with a reduction of IA presence and IA rupture in terms of SAH in the UK Biobank cohort.

Methods, Project duration:
Based on the UK Biobank cohort diet differences in participants without IAs, patients with unruptured IAs and patients with ruptured IAs shall be analysed mainly by t-test, non-parametric tests, correlation, logistic regression and cox regression. Analyses and manuscript preparation would take approximately 18-24 months.

Public health impact:
In most patients with unruptured IAs the risk for stroke and death in case of preventive repair outweighs the risk for rupture in case of observation. Conservative treatment options that even lower the rupture risk are urgently needed. Apart from this major argument a further argument for IAs with a low rupture risk would be the financial costs: preventive repair costs approximately 12,000-20,000 EUR (10,150-17,000 GBP), imaging for observation strategy approximately 500 EUR (425 GBP). If the intake of omega-3-fatty acids was associated with a lower rupture risk, this would be a promising conservative treatment option for patients with IAs and could lead to even fewer costs by imaging postponement.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-pain-killers-with-all-cause-and-cause-specific-mortality-among-patients-with-musculoskeletal-pain

Association of pain killers with all-cause and cause-specific mortality among patients with musculoskeletal pain

Last updated:
ID:
56837
Start date:
31 March 2020
Project status:
Current
Principal investigator:
Dr Lingxiao Chen
Lead institution:
University of Sydney, Australia

Pain killers (such as NSAIDs and opioids) are widely used in patients with musculoskeletal pain. But whether an association exists between pain killers’ usage and mortality is still debatable. Previous studies used old-fashioned methods, which might produce biased results. We will use a new analysis frame and try to give a more convincing conclusion. In additional, UK Biobank currently has the primary care data, which provides us an opportunity to identify the prescription trajectories (including the dosage and the frequency) for the pain killers. Thus, we could explore whether an association exists between prescription patterns of pain killers and mortality, which might guide the scientific usage of the pain killer to reduce the potential harm. In sum, we have three aims: 1. to determine whether an association exists between pain killers’ usage and mortality among patients with musculoskeletal pain; 2. to identify prescription trajectory patterns of pain killers among patients with musculoskeletal pain; 3.to explore whether an association exists between prescription patterns of pain killers and mortality among patients with musculoskeletal pain. We plan to finish this project in 15 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-physical-activity-fitness-and-obesity-with-cancer-incidence-and-mortality

Association of physical activity, fitness, and obesity with cancer incidence and mortality.

Last updated:
ID:
43456
Start date:
13 March 2019
Project status:
Current
Principal investigator:
Dr Charles Matthews
Lead institution:
National Cancer Institute, United States of America

While physical inactivity and obesity have been shown to increase risk of several cancers, current knowledge gaps exist due to the difficulty in obtaining reliable data from measures of physical activity (self-report) and body fat (body mass index) used in previous studies. By using more objective measures of both physical activity, fitness, and adiposity we can increase our understanding of how these behaviors are related to cancer risk. We propose to take advantage of the already available data from the UK biobank on physical activity (self-reported, accelerometer and cardiorespiratory fitness) and body fat (BMI and bioelectrical impedance) to address the previous limitations to assessing cancer risk. This project will provide further evidence on the roles physical activity and body fat have in cancer development and fill gaps in the literature regarding the importance of these modifiable risk factors for cancer prevention.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-psychiatric-disorders-with-subsequent-colorectal-cancer-prospective-cohort-study-of-uk-biobank-participants

Association of psychiatric disorders with subsequent colorectal cancer: prospective cohort study of UK Biobank participants

Last updated:
ID:
77512
Start date:
14 December 2021
Project status:
Current
Principal investigator:
Professor Jinlin Yang
Lead institution:
West China Hospital of Sichuan University, China

Colon cancer (CRC) is the third most common cancer in the world, with approximately more than 1.8 million new cases being reported annually. It is therefore important to identify risk factors for patient with CRC, with the aim to decrease the risk of CRC and reduce healthcare cost. Psychiatric disorders, especially depression and anxiety are common public health problem that has been associated with a number of chronic health outcomes, including cancers. Although certain psychiatric disorders (ie, depression) are established in the risk of CRC, previous studies on certain psychiatric problems and CRC incidence, most of which used moderate sample sizes or gender-limited samples, were inconsistent, reporting positive or null findings. To our knowledge, no comprehensive assessment of the role of multiple types of clinically confirmed pre-existing psychiatric disorders on CRC susceptibility has been done to date using longitudinal data. Therefore, this research aims to determine the association between psychiatric disorders and the subsequent risk of CRC. In this project, we are planning to finish the analysis of clinical data from UK Biobank in about 6 months, in order to find out the significant role of multiple types of psychiatric disorders in colorectal cancer, wish to provide more explicit information for clinical use and for psychological interventions, and to refined cancer prevention.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-risk-factors-in-terms-of-diet-lifestyle-genetic-susceptibility-metabolic-disorder-and-treatment-with-the-comorbidity-of-diabetes-with-cancer

Association of risk factors in terms of diet, lifestyle, genetic susceptibility, metabolic disorder and treatment with the comorbidity of diabetes with cancer.

Last updated:
ID:
90232
Start date:
27 July 2022
Project status:
Current
Principal investigator:
Professor Tianshu Han
Lead institution:
Harbin Medical University, China

Aims: this project aimed to identify the specific risk factors related to the comorbidity of diabetes and different types of cancer, to reveal their internal causal relationship and potential common mechanism based on plasma metabolites and genetics, and to establish the accurate evaluation model for cancer risk among diabetes for providing the personalized diabetes-cancer prevention and treatment strategies.
Scientific rationale: Previous cohort studies observed that cancer and diabetes was frequently coexisted. However, the association of diabetes and different types of cancer was heterogeneous, which was likely mediated by life-styles, diet, sex, genetics and micro-environment of hormonal and metabolic ability among different population. The previous evidence suggested that cancer and diabetes shared common mechanisms, such as inflammation, hyperinsulinemia, hyperglycemia, dysfunction of amino acids and fatty acids metabolism. Also, there are many studies have demonstrated that dietary risk factors, medication for diabetes or caner, and genetic risk factors could influence these above mechanisms. Based on these evidences, we hypothesized that susceptibility genes, physiological and pathological states, drug use could mediate the association of diabetes and different types of cancer, and elucidating these unravelling questions may added important knowledge for the intervention strategies for diabetes and cancer.
Project duration and public health impact: this project duration is estimated for 3 years. More than 60% of countries citing cancer as the top 1-2 cause of human death, which seriously threatens public health. Over 1/4 of people gets cancer during the lifetime, and over 17% of cancer patients have diabetes or abnormally plasma glucose. Previous studies reported that up to 10 types of cancer were attributed to obesity, hyperglycemia, and diabetes. Epidemiological studies showed that people with diabetes had a higher prevalence of malignant tumors, greater cancer process, poor treatment effect, and shortened survival time. Each 1% increase in the prevalence of diabetes was associated with 19% increase in the mortality rate of pancreatic cancer. According to a study in the Lancet, the per capita treatment cost for common cancers such as lung cancer and stomach cancer, is approximately $10,000 per year. Therefore, it is important to elucidate the potential risk factors for the cancer among patients with diabetes, which may improve the long-term survival among diabetes by providing evidence in terms of the prevention and treatment to reduce social financial burden on healthcare.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-risk-factors-in-terms-of-diet-lifestyle-genetic-susceptibility-metabolic-disorder-and-treatment-with-the-comorbidity-of-mental-disorder-and-cancer

Association of risk factors in terms of diet, lifestyle, genetic susceptibility, metabolic disorder and treatment with the comorbidity of mental disorder and cancer.

Last updated:
ID:
587642
Start date:
5 April 2025
Project status:
Current
Principal investigator:
Miss Yuling Ba
Lead institution:
Harbin Medical University Cancer Hospital, China

Research Questions:It has been observed that many cancer patients also suffer from mental disorders, yet the underlying mechanisms remain unclear. Lifestyle plays a significant role in either cancer or mental disorders. If the patterns within it can be identified, preventive measures could be developed. During the onset and progression of diseases, human blood biomarkers, such as blood metabolites and plasma proteins, often change. We aim to investigate whether such changes can more sensitively reflect disease onset and progression compared to clinical symptoms and develop predictive biomarkers or models. Diseases result from the combined effect of genes and the environment. Besides studying environmental and gene expression, we also intend to explore the genetic susceptibility of diseases and explain disease changes from a multi-omics perspective.
Objectives:This project aims to identify specific risk factors associated with the comorbidity of mental disorders and different types of cancer, uncover the intrinsic causal relationships and potential common mechanisms. Based on blood biomarkers and genetics, an accurate assessment model for the risk of mental disorders and cancer will be established to provide personalized cancer prevention and treatment strategies for patients with mental disorders.
Scientific Rationale:Large cohort studies have shown that patients with mental disorders have a higher risk of developing certain diseases than the general population. Cancer and mental disorders often coexist, and the comorbid health issues substantially increase the total cost of healthcare. However, the association between mental disorders and different types of cancer is heterogeneous, which may be mediated by factors such as gender, genetics, lifestyle, and the microenvironment of hormonal and metabolic capabilities in different populations. Previous evidence indicates that cancer and mental disorders share common mechanisms, such as inflammation and amino acid


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-sensory-impairment-disequilibrium-and-cognitive-frailty

Association of sensory impairment, disequilibrium and cognitive frailty

Last updated:
ID:
107217
Start date:
10 October 2023
Project status:
Current
Principal investigator:
Professor Haibo Wang
Lead institution:
Shandong Provincial ENT Hospital, China

Cognitive frailty is common in the elderly population. Sensory functions include taste, smell, touch, hearing, and sight, as well as spatial orientation and ability of balance. Sensory function and equilibrium declines with age. In turn, sensory impairments and disequilibrium predicts accelerated cognitive aging. However, There remains unclear the relationship between sensory impairment, disequilibrium and cognitive frailty.
The current research aims to 1) understand how sensory impairment, disequilibrium relate to cognitive function/frailty; 2) find human fluid, blood and multiple omics indicators can be used to indicate cognitive frailty, explore the potential mechanisms that may explain this relationship; 3)reveal the information on any intervention to improve sensory function and balance ability that can prevent people from cognitive frailty. The UK Biobank data will be particularly useful in terms of providing brain structure and function, clinical biomarkers for understanding sensory and balance impairment in cognitive frailty. Findings will provide a better understanding of potential influencing of sensory input on cognitive frailty, and will highlight the importance of intervening in different sensory systems to Slow or reverse cognitive decline.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-serum-calcium-vitamin-d-and-c-reactive-protein-with-all-cause-and-cause-specific-mortality-in-osteoarthritis

Association of serum calcium, vitamin D and C-reactive protein with all-cause and cause-specific mortality in osteoarthritis

Last updated:
ID:
69521
Start date:
6 April 2022
Project status:
Current
Principal investigator:
Dr Kai Fu
Lead institution:
Shanghai Sixth People's Hospital, China

Osteoarthritis is the most common musculoskeletal disease and the leading cause of disability globally. Vitamin D deficiency is the most common nutritional deficiency worldwide which can also regulate the serum calcium. In addition, the serum C-reactive protein was also highly associated with osteoarthritis. However, there are few studies investigating the effect of serum calcium, vitamin D and C-reactive protein on the all-cause and cause-specific mortality in osteoarthritis population.
Our project will involve looking at the baseline serum calcium, vitamin D and C-reactive protein and their associations with mortality in the osteoarthritis population. We expect the project duration to be a period of several months. This research will help us to understand more about the role of serum calcium, vitamin D and C-reactive protein in osteoarthritis, in particular, their longitudinal roles in death.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-serum-markers-lifestyle-and-health-outcomes-exploring-risk-factors-for-cardiovascular-and-cerebrovascular-diseases-and-mortality

Association of Serum Markers, Lifestyle, and Health Outcomes: Exploring Risk Factors for Cardiovascular and Cerebrovascular Diseases and Mortality

Last updated:
ID:
106487
Start date:
29 June 2023
Project status:
Current
Principal investigator:
Professor Wen Sun
Lead institution:
University of Science and Technology of China, China

Aims: Our research project aims to investigate the relationship between serum markers, lifestyle factors, and the risk of cardiovascular and cerebrovascular diseases, as well as all-cause mortality. We also aim to explore the genetic contributions to these health outcomes. By understanding these associations, we hope to identify potential risk factors and improve our understanding of the underlying mechanisms.

Scientific Rationale: Cardiovascular and cerebrovascular diseases, such as heart disease and stroke, are leading causes of death worldwide. Lifestyle factors and serum markers, which are measurable substances in the blood, have been linked to the development of these diseases. By studying a large cohort from the UK Biobank database, we can explore how lifestyle choices and serum markers influence disease risk and mortality. Additionally, we will investigate the genetic components that may contribute to these outcomes.

Project Duration: The research project is expected to be conducted over 3 years. The exact timeline will be determined based on the complexity of the research questions and the availability of the required data.

Public Health Impact: The findings of this study have the potential to significantly impact public health. By identifying specific lifestyle factors and serum markers associated with cardiovascular and cerebrovascular diseases, we can develop targeted interventions and strategies for disease prevention and management. Furthermore, understanding the genetic contributions to these health outcomes can lead to the development of personalized approaches to healthcare, enabling individuals to make informed decisions about their health based on their genetic risk profiles.

Overall, our research project aims to shed light on the associations between serum markers, lifestyle factors, cardiovascular and cerebrovascular diseases, and all-cause mortality. By leveraging the UK Biobank database, we hope to uncover valuable insights that can inform public health policies, improve risk prediction models, and ultimately contribute to reducing the burden of these diseases in the population.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-social-and-environmental-factors-and-genetic-markers-with-the-risk-of-postpartum-depression-in-women

Association of social and environmental factors and genetic markers with the risk of postpartum depression in women.

Last updated:
ID:
86155
Start date:
8 February 2023
Project status:
Current
Principal investigator:
Ms Rachel Kenny
Lead institution:
Indiana University Purdue University Indianapolis, United States of America

This proect aims to assess the association between post-partum depression (PPD) and various social, environmental, and genetic risk factors. Between 10% and 15% of women who give birth will experience PPD which includes all symptoms common in clinical depression and several specific to the postpartum experience. PPD is of special clinical significance due to it’s potential to affect not only the new mother but also the newborn. One area of ongoing research for postpartum depression is in genetics as prior research has shown those with a family history of depression have an increased risk of developing PPD. This finding suggests a possible genetic proponent which could be used to identify women with a genetic prediposition for developing PPD before it occurs. This project aims to advance the understanding of risk factors for PPD and will enable physicians to better predict women most at risk for developing this disease. It is estimated that this project will be completed within 18 months upon receipt of the data.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-the-interaction-between-metabolic-risk-factors-and-smoking-with-the-risk-of-lung-cancer

Association of the interaction between metabolic risk factors and smoking with the risk of lung cancer.

Last updated:
ID:
104126
Start date:
19 July 2023
Project status:
Current
Principal investigator:
Dr Hongxia Li
Lead institution:
The First Affiliated Hospital, Sun Yat-sen University, China

In this project, we will identify the potential interaction between metabolic profiles and smoking in the development and outcomes of lung cancer. Metabolic factors will be investigated, including detailed alcohol intake, body mass index (BMI), waist circumference, blood pressure, diabetes, consumption of fruit and vegetables, supplementation of nutrients and medication, and blood biochemistry (triglycerides, HDL-cholesterol, C-reactive protein et al). Cigarette smoking contributes the most to lung cancer occurrence and mortality. Despite recent advances, lung cancer therapies have shown limited clinical benefit. Given the need for new therapies, the metabolism of lung cancer has been widely studied in the past two decades to identify vulnerabilities that could be translated into novel anti-metabolic therapeutic approaches. Emerging evidence has highlighted the role of glucose, glutamine, and mitochondrial metabolism in the development of lung cancer. Therefore, both smoking and metabolic factors affect the development of lung cancer. However, whether metabolic factors interact with smoking to promote lung cancer development is still unclear, and their combined effects must be considered. The project is planning to take 3 years after the data has been downloaded. By linking metabolic risk factors to smoking-promoted lung cancer development, it is possible to identify people and populations that are at higher risk and offer early intervention. Accordingly, this deeper understanding of lung cancer may contribute to improving diagnosis, prevention and treatment strategies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-the-life-pattern-with-the-risk-of-myocardial-infarction-mi-in-people-without-standard-modifiable-cardiovascular-risk-factors-smurfs

Association of the life pattern with the risk of myocardial infarction (MI) in people without standard modifiable cardiovascular risk factors (SMuRFs)

Last updated:
ID:
97155
Start date:
7 March 2023
Project status:
Current
Principal investigator:
Dr Yanjun Song
Lead institution:
Chinese Academy of Medical Sciences &Peking Union Medical College, China

SMuRFs, which include hypertension, dyslipidemia, diabetes mellitus and smoking, are demonstrated to increase the risk of cardiovascular events with no doubts. However, it has been found that a non-negligible proportion of patients with MI combined no SMuRF (13-27%), and the risk of people with no SMuRF is increasing for each year (2004-2019: 11-27%). Therefore, people with no SMuRF, who were described as SMuRF-less status, attached great attentions, and potential reasons for this phenomenon are still not elucidated. Previous studies have demonstrated that unhealthy lifestyles such as unhealthy diet and bad sleep pattern are significantly associated with increased incidence of cardiovascular events. Therefore, it raises great interests that whether the unhealthy life pattern is a pivotal contributor to MI risk in people with SMuRF-less status, which still lacks of explorations. In this investigation, life pattern was evaluated from 7 dimensions: diet, alcohol, sleep pattern, environmental exposure, socioeconomic status and mental health, and the association of different healthy extent of the life pattern with the risk of MI will be investigated in people without SMuRFs. Besides, SMuRF-less people with healthy or unhealthy life pattern will be compared with people with SMuRFs in the risk of MI, further determine the concrete risk brought by unhealthy life patterns in SMuRF-less people. Finally, we will document that whether the unhealthy life pattern is a key point which will bring huge risk of MI and should be paid more attentions in SMuRF-less patients.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-the-metabolic-syndrome-and-its-components-with-bladder-cancer-an-exploratory-factor-analysis

Association of the metabolic syndrome and its components with bladder cancer! an exploratory factor analysis

Last updated:
ID:
206552
Start date:
3 October 2024
Project status:
Current
Principal investigator:
Dr Weiming Liang
Lead institution:
Guangxi University of Science and Technology, China

Metabolic syndrome (MetS) is a complex clinical syndrome characterized by obesity, hypertension, hyperglycemia, dyslipidemia, and other metabolic abnormalities. Insulin resistance is the central feature of this syndrome. Irrespective of the specific definition of MetS, its occurrence is on the rise globally. In countries where obesity rates are high and dietary habits are unhealthy, MetS has become a prevalent medical condition and a major public health concern, resulting in substantial economic burdens. Research has shown that MetS is associated with an elevated risk of bladder cancer. Additionally, individuals with MetS tend to have bladder cancer that is more advanced in terms of pathological grade and stage. There is a scarcity of evidence about the correlation between bladder cancer patients with MetS and their oncological outcomes, such as death, overall survival, cancer-specific survival, disease recurrence, and disease progression. We plan to utilize various data sources, including the UK Biobank and the Alpha Omega cohort, to conduct a more comprehensive analysis of these connections. The aim of our study was to thoroughly assess the association between each metabolic component of Metabolic Syndrome (e.g., obesity, diabetes) and the likelihood of developing bladder cancer or experiencing oncologic outcomes. Specifically, we also aim to examine whether the collective impact of MetS is higher or lesser than the single effect of its individual metabolic components. Our results are expected to provide a theoretical basis for the prevention, diagnosis and treatment of bladder cancer.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-triglyceride-glucose-tyg-index-with-incidence-of-hypertension

Association of triglyceride-glucose (TyG) index with incidence of hypertension

Last updated:
ID:
73887
Start date:
7 March 2022
Project status:
Current
Principal investigator:
Dr Javad Alizargar
Lead institution:
National Taipei University of Nursing and Health Sciences, Taiwan, Province of China

Aims: To know if different levels of triglyceride-glucose (TyG) index is associated with incidence of hypertension in individuals.
scientific rationale: Insulin resistance has been recognized to play a significant role in many diseases that involve heart and vessels. TyG index is a marker of insulin resistance. As insulin resistance is a predictor of diabetes and diabetes has a high association with new onset hypertension, TyG might be an independent predictor of new onset hypertension.
project duration: After getting access to the database, data cleaning based on inclusion and exclusion criteria, final data will be prepared for analysis (1 month). Data analysis by our data scientists consists of general categorization and simple statistics followed by advanced statistical analysis (2 months). After getting the main results preparing the report and preparation of the manuscript to submit to scientific journals (2 months).
public health impact: By knowing if the insulin resistance index can be an independent factor for development of hypertension, we can set cut of values to warn people at risk and improve the adverse health outcome of hypertension.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-type-2-diabetes-according-to-the-number-of-risk-factors-within-target-range-with-risk-of-depression

Association of type 2 diabetes, according to the number of risk factors within target range, with risk of depression

Last updated:
ID:
81410
Start date:
22 February 2022
Project status:
Closed
Principal investigator:
Dr Thomas van Sloten
Lead institution:
Maastricht University Medical Center, Netherlands

Individuals with diabetes have a higher risk of developing depression compared to individuals without diabetes. The extent to which this excess risk may be reduced by current treatment is unclear. Current treatment consists of targeting multiple factors, including high blood sugar levels, elevated blood pressure, being overweight, smoking and elevated protein levels in urine. In this project, we will evaluate to which extent the risk of depression in diabetes may be potentially mitigated by this multifactorial treatment. This will inform patients and medical doctors about the potential beneficial effects of current treatments. The estimated project duration is 12 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-ultraprocessed-food-upf-consumption-with-risk-of-age-related-macular-degeneration-amd

Association of ultraprocessed Food (UPF) consumption with risk of age related macular degeneration (AMD)

Last updated:
ID:
104951
Start date:
22 June 2023
Project status:
Current
Principal investigator:
Professor Ai Zhuang
Lead institution:
Shanghai Ninth People's Hospital, China

There has been a growing body of evidence associating consumption of ultra-processed foods (UPF) with adverse health outcomes including depression, cardiovascular disease, dementia, and all-cause mortality. Age-elated macular degeneration (AMD) is a typical ocular vascular and aging disease lead to severe blindness. However, whether UPF are associated with AMD is unknown. We aim to investigate the associations between UPF and AMD incidence in UK biobank. This study will last three years. Finally, this study will add knowledge whether UPF are associated with AMD, and give people at risk of AMD a healthy diet.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-of-vitamin-d-and-tea-comsumption-wth-alzheimer-disease

Association of Vitamin D and tea comsumption wth Alzheimer disease

Last updated:
ID:
88813
Start date:
6 July 2022
Project status:
Current
Principal investigator:
Yin Wang
Lead institution:
Central South University, China

The prevalence of dementia is increasing and diet as a modifiable factor could play a role.Alzheimer’s disease (AD), the main cause of dementia in older adults,is a significant worldwide concern because of its high prevalence and incidence, adverse consequences, and lack of curative medications.Accumulating evidence has underlined that adverse effects of Vitamin D are not limited to the bone but, depending on the serum level of 25-hydroxyvitamin D (25OHD), also affect the brain. Evidence on the role of vitamin D in the central nervous system has increasingly been described.
Previous studies have revealed the involvement of tea in the development of dementia. However, little is known about the association between the combination of tea and the risk of AD. Therefore, my study will aim to investigate the association of vitamin D and tea separately with the risk of AD.
Patients with AD suffer from a lot and usually cost so much with threapy.My study, which may last half a year, aim to investigate the association of Vitamin D and tea consumption with Altheimer disease, so that the diseae could be prevented on diet.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-studies-for-allergies-and-autoimmune-diseases

Association Studies for Allergies and Autoimmune Diseases

Last updated:
ID:
225316
Start date:
2 April 2025
Project status:
Current
Principal investigator:
Professor Joshua Milner
Lead institution:
Columbia University, United States of America

Existing research with genetic data has begun to uncover many genes that are commonly found in patients with diseases related to allergies and the immune system. However, this type of research uncovers many more genes that can possibly cause these disorders – some uncovered genes cause these disorders, but others are simply likely to be found alongside! This phenomenon (known as linkage disequilibrium), that having one gene can make someone more likely to have another unrelated gene, makes it hard to identify which gene is actually causing the studied diseases. This research will focus on using additional data from donors, especially data concerning genes that have already been turned into proteins, to help narrow down from “associated genes” to “causal genes”.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-studies-of-infertility-cardiovascular-diseases-and-type-2-diabetes

Association Studies of Infertility, Cardiovascular Diseases, and Type 2 Diabetes

Last updated:
ID:
33672
Start date:
15 June 2017
Project status:
Closed
Principal investigator:
Professor Rasmus Nielsen
Lead institution:
University of California, Berkeley, United States of America

We aim to verify various disease associations we see in isolated populations to those in UK Biobank. The main research problems we plan to investigate are:

1) Do the genetic variants associated with type 2 diabetes and cardiovascular diseases in the Andean and the Inuit populations also show a significant association in the UK Biobank samples?

2) Do the genetic variants associated with infertility in Chinese populations also show a significant association in the UK Biobank samples?

3) How do certain phenotypes interact with each other? How do we best control for interactions among phenotypes in association studies? Identification of genetic variants related to type 2 diabetes, cardiovascular diseases, or infertility will help researchers develop better methods to detect and target these conditions for treatment. Furthermore, studying differences in disease associations among different populations will help researchers develop population-specific methods in detecting, preventing, and treating conditions of interest. We will conduct genome-wide association studies on the UK Biobank samples to identify genetic variants associated with phenotypes related to type 2 diabetes, infertility, and cardiovascular diseases. We will compare the results to the variants identified in the Andean, Inuit, and Chinese populations to verify the associations found in these populations and investigate potential differences between populations.

We will also conduct a meta-analysis to study the interactions between different phenotypes, which would help us define and control interacting terms in our associations studies. We would like to request access to the full cohort.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-studies-of-tandemly-repeated-sequences-in-the-uk-biobank

Association studies of tandemly repeated sequences in the UK Biobank

Last updated:
ID:
82094
Start date:
18 February 2022
Project status:
Current
Principal investigator:
Dr Andrew James Sharp
Lead institution:
Icahn School of Medicine at Mount Sinai, United States of America

Contrary to the commonly held belief that humans carry two copies of each gene, the human genome actually contains many coding regions and entire genes that occur in multiple copies. These “multicopy genes” have been largely ignored by most genetic studies, in part because they are inherently very difficult to study. However, they also often vary in copy number between different people, and this copy number variation can alter genetic risks for disease. We have developed new methods to study these multicopy genes. Here, we propose to investigate how people that carry different numbers of these multicopy genes might influence their risk of getting many different diseases. Over the next 3 years, we hope to identify variation of which multicopy genes modify our disease risks, which we hope will lead to new insights a drug targets that might ultimately improve human health.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-study-between-adenomyosis-and-preterm-birth

Association study between adenomyosis and preterm birth

Last updated:
ID:
211468
Start date:
25 November 2024
Project status:
Current
Principal investigator:
Ms Meng LiLi
Lead institution:
Guangdong Second Provincial General Hospital, China

Our research aims to understand why some women with adenomyosis, a condition where the lining of the uterus grows into the muscle, are more likely to have premature births. We’ll use data from the UK Biobank to study the genes of thousands of women with adenomyosis and premature births. By doing this, we hope to find out if there are specific genes that increase the risk of both conditions. Understanding this could help us develop better ways to prevent and treat these problems.

The reason we’re interested in this is because both adenomyosis and premature birth are big concerns for women’s health. Adenomyosis can cause painful periods and heavy bleeding, while premature birth can lead to serious health problems for babies. We know that there’s a genetic component to both conditions, but we don’t fully understand how they’re linked. By studying the genes of women with adenomyosis who also had premature births, we can try to unravel this connection.

Our project will last for about three years. During this time, we’ll analyze the genetic data of thousands of women to look for patterns that might explain why some women with adenomyosis have premature births. We’ll also compare our findings with women who don’t have these conditions to see if there are differences in their genes.

The impact of our research could be significant. By identifying the genes involved in both adenomyosis and premature birth, we could pave the way for new treatments or preventive measures. This could improve the quality of life for women with adenomyosis and reduce the number of premature births, benefiting both mothers and babies. Ultimately, our goal is to contribute to better reproductive health outcomes for women around the world.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-study-between-diabetes-and-bone-mineral-density-based-on-uk-biobank-data-plasma-proteomics-analysis-and-causal-exploration

Association Study between Diabetes and Bone Mineral Density Based on UK Biobank Data: Plasma Proteomics Analysis and Causal Exploration

Last updated:
ID:
863859
Start date:
22 July 2025
Project status:
Current
Principal investigator:
Miss Ying Xue
Lead institution:
North China University of Science and Technology, China

Research Questions:

What is the longitudinal association between plasma protein levels and osteoporosis risk in diabetic patients?

Which proteins are causally linked to bone mineral density (BMD) changes in diabetes, and what are their functional roles?

Can a predictive model integrating proteomic data improve osteoporosis risk stratification in diabetes compared to traditional clinical factors?

Objectives:

Identify plasma proteins associated with BMD changes in diabetic individuals using UK Biobank data.

Explore causal relationships via Mendelian Randomization (MR) leveraging GWAS and pQTLs.

Develop a machine learning model to predict osteoporosis risk based on proteomic signatures.

Investigate biological pathways of candidate proteins through network and enrichment analyses.

Scientific Rationale:

Diabetes mellitus (types 1 and 2) is linked to altered bone metabolism, but the underlying mechanisms remain unclear. Plasma proteomics offers a systemic view of protein biomarkers influencing bone health. This study leverages the UK Biobank’s large-scale proteomic (Olink) and DXA-derived BMD data to:

Address gaps by longitudinally assessing protein-BMD associations while adjusting for confounders (e.g., age, BMI).

Elucidate causality using MR to mitigate confounding biases inherent in observational studies.

Translate findings into clinical tools (e.g., predictive models) for early osteoporosis intervention in diabetes.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-study-of-rare-metabolic-disorders-and-common-metabolic-syndromes

Association study of rare metabolic disorders and common metabolic syndromes

Last updated:
ID:
70726
Start date:
17 May 2021
Project status:
Current
Principal investigator:
Professor Xiling Shen
Lead institution:
Duke University, United States of America

Obesity and associated metabolic syndromes are global health issues threatening our society. Ample evidence shows calories from different nutrients are not equal and could drive the development of metabolic syndromes. While metabolomics gains popularity, the effect of metabolites in both bloodstream and tissues remain controversial. This study lavage the abnormal metabolite level seen in a population harbors rare mutation to investigate the effect of different metabolites. By restricting the analysis to metabolism-related genes, we expect to unveil many previously overlooked associations between the loci and clinical outcomes. In approximately thirty months, we will shortlist metabolites that are likely associated with different metabolic syndromes. These metabolites can be used for disease monitoring and served as drug targets.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/association-tests-for-structured-and-multimodal-data

Association tests for structured and multimodal data

Last updated:
ID:
40502
Start date:
1 August 2018
Project status:
Closed
Principal investigator:
Professor Christoph Lippert
Lead institution:
Max Delbruck Center for Molecular Medicine, Germany

The goal of this project is to develop new methods that help biomedical scientists in the analysis of their data.
Technological advances in clinical measurement devices based on sequencing, imaging, and wearables promise to accurately diagnose diseases more accurate and in their earliest stages when they can be readily treated.
Machine learning is central to this vision of personalized medicine, where each individual is monitored based on their medical history, as well as their own genetic and environmental disease risk.
With the size and complexity of the data mounting, computational and statistical analysis is becoming a bottleneck.
We develop new methods from artificial intelligence and machine learning to analyze large volumes of complex data that are derived from clinical measurements and derive new scientific knowledge in an increasingly automated fashion and aid the medical experts in data interpretation.
In order to enable us to interpret the disease risk of an individual, we need to develop models that view the risk of an individual relative to a healthy population, and relative to individuals who have a disease.

This project not only focuses on methods development in machine learning (ML) and statistics to make this vision become a reality, but also aims on developing such models that ultimately will serve as an empirical footing for personalized preventative medicine.
Accurate statistical models as we plan to develop that take into account confounding are essential to determine robust associations and to derive precise risk models for diseases in the presence of environment, lifestyle, medication, and molecular measurements.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-among-24-hour-movement-behaviours-and-markers-of-physical-and-mental-health-cognitive-and-physical-functioning-metabolic-risk-and-disease-status-a-compositional-data-analysis-approach

Associations among 24-hour movement behaviours and markers of physical and mental health, cognitive and physical functioning, metabolic risk, and disease status: A compositional data analysis approach

Last updated:
ID:
64028
Start date:
25 January 2021
Project status:
Closed
Principal investigator:
Professor Martyn Standage
Lead institution:
University of Bath, Great Britain

Each day, people engage in the behaviours of sleep, sedentary time, light physical activity, and moderate-to-vigorous physical activity. Separately, these behaviours have been shown to predict a number of health outcomes such as a person’s mental health, their risk of disease, their physical health, and their functional ability. However, the analyses conducted previously do not account for the fact that over a 24-hour period (or 1440 minutes), a change in the amount of one behaviour naturally results in changes to the other behaviours. In this work, an analysis that captures collective 24-hour movement behaviour data appropriately will be used to see how different daily patterns relate to important health outcomes (i.e., essentially, “what makes for a healthy or unhealthy day”). These outcomes include measures of mental health such as depression and anxiety, subjective well-being (e.g., mood and happiness), cognitive function (e.g., memory, reasoning, reaction time), physical functioning (e.g., handgrip strength, blood pressure, body size measurements), metabolic risk (i.e., a combination of factors that predict important outcomes such coronary heart disease and stroke), and disease status (e.g., having heart disease, diabetes, cancer, etc). In addition to academic impact, it is expected that the results of this project will inform public health policy and practice.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-and-mechanisms-linking-transport-and-health

Associations and mechanisms linking transport and health

Last updated:
ID:
89121
Start date:
8 September 2022
Project status:
Current
Principal investigator:
Dr Sandar Tin Tin
Lead institution:
University of Auckland, New Zealand

Regular physical activity is widely recommended to promote health and to prevent a range of health conditions. However, many exercise programmes require special skill, time or expense, discouraging sustained participation particularly by disadvantaged groups. One practical solution would be to build physical activity into the daily routine by using active modes of transport. Active travel has been shown to improve health and also provide social, economic and environmental benefits. However, little is known about its effects on more specific health conditions. Car use, on the other hand, is a significant contributor of daily sedentary time but its effects on health is not well understood. This research therefore aims to investigate if and how transport choices influence physical, mental and cognitive health, and site-specific cancers. The findings will help tailor health promotion strategies for intervention and improve public health guidelines. There will also be opportunities for post-graduate student training in longitudinal research and big data analysis.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-between-air-pollution-and-respiratory-health-in-large-european-cohorts

Associations between air pollution and respiratory health in large European cohorts

Last updated:
ID:
9946
Start date:
31 December 2014
Project status:
Closed
Principal investigator:
Dany Doiron
Lead institution:
Research Institute McGill University Health Centre, Canada

This project will examine associations between exposure to common ambient air pollutants and respiratory health among participants of large population-based studies in the Netherlands and the United Kingdom. Research questions for this project are:
(1) What is the effect of exposure to particulate matter (PM) and nitrogen dioxide (NO2) on pulmonary function?
(2) Do study participants living in areas with higher concentrations of particulate matter (PM) and nitrogen dioxide (NO2) show higher rates of respiratory medication use and respiratory symptoms such as wheezing and shortness of breath? There is mounting evidence showing associations between air pollution exposure and adverse cardiovascular and respiratory health effects. However, associations between common air pollutants such as particulate matter and nitrogen dioxdide and pulmonary function, respiratory symptoms and respiratory medication use are not well understood. Research is therefore needed to better understand the effect that air pollution has (if any) on respiratory health and pulmonary function decline. Better understanding these effects is in the public interest given the high health care costs related to respiratory morbidity and mortality. This project involves harmonizing and combining data from two of Europe?s largest population health studies, the LifeLines Cohort Study & Biobank and UK Biobank, in order to:
(1) Explore associations between PM/NO2 exposure at place of residence and pulmonary function and,
(2) Explore the effect of PM/NO2 exposure at place of residence on prevalence respiratory symptoms (e.g. wheezing and shortness of breath) and respiratory medication use.

Pulmonary function, medication use, respiratory symptoms and confounding variables will be obtained from baseline assessments. Modeled air pollution measures at place of residence will be linked to these data for analyses. All UK Biobank participants 18 years and older having completed the pulmonary function test and for which data requested has been cleaned and validated should be included in the dataset.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-between-air-quality-regional-brain-volumes-cognitive-functioning-and-neuropsychiatric-outcome-in-middle-aged-and-older-adults-a-cross-sectional-study

Associations between air quality, regional brain volumes, cognitive functioning, and neuropsychiatric outcome in middle-aged and older adults: a cross-sectional study

Last updated:
ID:
41535
Start date:
6 August 2018
Project status:
Current
Principal investigator:
Dr Dawson Ward Hedges
Lead institution:
Brigham Young University, United States of America

In this study, we aim to identify and elucidate associations in adults between exposure to air pollution and cognitive function, mood disorders, and brain structure.

Exposure to air pollution has been associated with increased risk for respiratory and cardiovascular disease, and recent work suggests an association between exposure to air pollution and worsened cognitive function in children and adults and possibly dementia. In that air pollution has been associated with cognitive function, we hypothesize that it might be associated with the mood disorders depression and bipolar disorder. In addition, air pollution has been associated with abnormal brain volume in children and adults. Air pollution might be associated with cognitive function via damage to the protective blood-brain barrier or inflammation. We propose to use data from the UK Biobank to investigate further the relationship between exposure to air pollution and cognitive and mood outcomes and brain volumes. To identify factors that might influence any associations between air pollution and cognition, mood disorders, and brain volume, we plan to investigate the potential effects of several medical and social variables, such as age, sex, and educational attainment.

If this application is successful, we anticipate completing the project in six months after receiving the data.

Given the numbers of people including children worldwide exposed to air pollution and findings showing possible associations between exposure to air pollution and cognitive development, cognitive function, and brain volume, additional information characterizing these associations in large, controlled studies are vital for public interest and global health, particularly considering increasing global urbanization and subsequent exposure to air pollution. Identification of risk and protective factors is also important in better understanding the effects of air pollution on cognitive and mental health.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-between-body-compositions-with-the-development-of-cardiometabolic-diseases-and-cognitive-disorders

Associations between body compositions with the development of cardiometabolic diseases and cognitive disorders

Last updated:
ID:
213095
Start date:
9 January 2025
Project status:
Current
Principal investigator:
Professor Cheng Hu
Lead institution:
Shanghai Jiao Tong University, China

Cardiometabolic diseases (CMDs) and cognitive disorders, including cardiovascular diseases, type 2 diabetes, and Alzheimer’s disease, stand as prominent global causes of morbidity and mortality, imposing substantial burdens on individuals and society. Therefore, there is a critical need to identify early risk predictors and potential therapeutic targets to mitigate the impact of these diseases.
Body compositions, comprising adipose tissue, skeletal muscle, and bone, are recognized as the body’s primary metabolic organs. Previous findings from in vivo and in vitro experiments suggest that these body compositions can secrete various cytokines involved in disease development and organ cross-talk. However, the roles of different body compositions in the development of CMDs and cognitive disorders in humans remain largely unexplored due to the lack of data from large-scale population-level studies.
The project is set to span 36 months, utilizing high-quality data from the UK Biobank (UKB). Our aim is to identify features of body compositions that can effectively predict the development of CMDs and cognitive disorders. These insights may serve as early risk indicators, enabling the identification of individuals at a heightened risk for these diseases. Additionally, the study seeks to identify potential therapeutic targets for related diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-between-chronic-obstructive-pulmonary-disease-measures-and-time-use

Associations between chronic obstructive pulmonary disease measures and time use

Last updated:
ID:
28231
Start date:
30 June 2017
Project status:
Closed
Principal investigator:
Ms Hayley Lewthwaite
Lead institution:
University of South Australia, Australia

Chronic obstructive pulmonary disease (COPD) is characterised by chronic, progressive airflow limitation. Severity of airflow limitation and presence of comorbidities contribute to overall severity, increasing risk of all-cause mortality and morbidity. Physical activity, sedentary behaviour and sleep (time-use) may play a role in mitigating the effects of COPD.

This research project aims to address two questions:
1) Is there an association between COPD status and time-use?
2) Does time-use modify the relationship between COPD status and all-cause mortality and hospitalisation?
While COPD is not curable, early diagnosis and effective management can slow disease progression, reducing associated mortality and morbidity. Worldwide, 8-15% of adults =40 years have COPD; causing an estimated 3 million deaths/year and contributing to years lived with disability. A key component of COPD management is optimising function, which pulmonary rehabilitation (PR) plays an important role. While PR has shown to improve symptoms and function, the benefits don?t last long-term. Targeting daily time-use may be a more effective strategy for long-term management. Further research is needed to explore associated health benefits in COPD. Statistical analyses will be used to explore associations between COPD status (composite score likely to include spirometry measures, breathlessness and COPD-hospital admission frequency) and physical activity, sedentary behaviour and sleep (accelerometer data and/or self-report measures). A previously defined COPD-specific index will be used (COTE) to quantify comorbidity status, which will be used as a covariate.

The second research question will involve exploring the relationship between COPD status and incidence of hospitalisation and death, and then assessing whether physical activity, sedentary behaviour and sleep modify this relationship
Participants who have spirometry data will be sought (n=353,403). Data over all instances (0-2) will be obtained.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-between-covid-19-symptoms-stressful-life-experiences

Associations between COVID-19 Symptoms & Stressful Life Experiences

Last updated:
ID:
92699
Start date:
16 September 2022
Project status:
Current
Principal investigator:
Dr Jamie Lars Hanson
Lead institution:
University of Pittsburgh, United States of America

This project will examine the relationship between stress, stressful life experiences, and COVID-19. Stress is a risk factor for many illnesses, including infectious diseases such as viruses. Stress can increase both the likelihood of developing an illness and the severity of symptoms. Preliminary research suggests that childhood trauma, a stressful life experience, increases the likelihood of developing “long COVID”, a COVID infection with symptoms lasting 3+ months. In our research, we will determine if stress and stressful life experiences are significant risk factors for severe COVID illness.

To conduct our research, we will look at patients with positive COVID-19 diagnoses and examine medical records to determine symptoms. We will look at a variety of metrics related to mental health, including current stress levels and recent stressful experiences, as well as stressful experiences that happened in childhood. We will also consider medical diagnoses, such as diabetes, and sociodemographic factors, such as age, as potential confounding variables. We will use multiple measurements to determine severity of COVID illness: severity of symptoms, total number of symptoms, and duration of symptoms. Using this data, we will investigate if more severe illness correlates with stress and stressful life experiences.

As the data is already collected, we aim to complete our analyses within the span of approximately 18 months. Regardless of our findings, this study will have a positive impact on public health. If we determine that stress and stressful life experiences are risk factors for severe illness, this will help treatment and prevention at multiple levels. Individuals will be able to more accurately assess their own risk and make informed decisions regarding their health and safety. Doctors will be able to better predict if their patients will develop severe illness, and create treatment plans accordingly. Public health institutions will be able to target at-risk populations for testing and prevention efforts. If we determine that stress and stressful life experiences are not risk factors for severe illness, future researchers can focus on investigating other potential risk factors.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-between-cxcr4-genetic-variants-and-immunodeficiency-phenotypes

Associations between CXCR4 genetic variants and immunodeficiency phenotypes

Last updated:
ID:
81315
Start date:
8 February 2022
Project status:
Closed
Principal investigator:
Dr Katarina Zmajkovicova
Lead institution:
X4 Pharmaceuticals, Austria

Primary immunodeficiencies (PIDs) are a group of inherited diseases that result in impairment of normal immune system function. The affected individuals often suffer from recurrent infections. Warts, Hypogammaglobulinemia, Infections and Myelokathexis (WHIM) syndrome, one such PID, causes different symptoms including low immune cell counts in blood, bacterial infections or skin warts and it is caused by mutations in the C-X-C chemokine receptor type 4 (CXCR4) gene. Currently there is a small number of pathogenic mutations in CXCR4 that are known to cause WHIM syndrome. Novel CXCR4 mutations are sometimes identified in patients with immunodeficiency, but due to missing information on their pathogenicity, the diagnosis of WHIM syndrome cannot be confirmed. As a result, these individuals may not be eligible for the best suitable treatment. We plan to look at a large group of people in the UK Biobank to identify individuals who have CXCR4 mutations as well as low blood cell counts or other signs of immunodeficiency. We will analyze these mutations in laboratory tests and then combine all data to conclude which of the mutations are likely to cause decreased immune system function. This study will form the foundations for better diagnosis of WHIM syndrome or other CXCR4-related immunodeficiency. The project will last for one year.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-between-diet-body-composition-and-co-morbidities-in-uk-biobank-participants-with-psoriasis

Associations between diet, body composition, and co-morbidities in UK Biobank participants with psoriasis

Last updated:
ID:
88354
Start date:
14 July 2022
Project status:
Current
Principal investigator:
Dr Wendy Louise Hall
Lead institution:
King's College London, Great Britain

Psoriasis is an immune related skin disorder that affects around 2-3% of the UK population, with adverse psychological, physical and social consequences. Psoriasis is associated with many diet-related co-morbidities such as metabolic syndrome, type 2 diabetes, cardiovascular disease (CVD) and psoriatic arthritis. Little is known about the role of diet in the causation and management of psoriasis. We will characterise the diets of UK Biobank participants with psoriasis and test whether diet quality is associated with a lower risk of psoriasis.
Aims: The primary aim of our research is to characterise the diets of people living with psoriasis, determine whether diet quality is associated with psoriasis comorbidities and medications, and whether this relationship varies among participant subgroups (i.e. those with overweight/obesity, demographic categories such as socioeconomic status, co-morbidities such as digestive disorders). Finally, we will examine the relationship between dietary factors and incidence of psoriasis.
Scientific rationale: Psoriasis is associated with both overweight/obesity and inflammation, both of which are influenced by diet quality. In view of the fact that common co-morbidities are diet-dependent (CVD, gastrointestinal diseases, diabetes), it is important to characterise the diets of those with psoriasis, and to investigate whether low adherence to healthy dietary patterns is associated with psoriasis and co-morbidities.
Project duration: This project will be conducted over 3 years as part of a PhD project.
Public Health Impact: If we find an association between diet quality and psoriasis, future randomised controlled trials can be used to investigate efficacy of dietary interventions in reducing disease severity.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-between-environmental-factors-and-multiple-health-outcomes

Associations between environmental factors and multiple health outcomes

Last updated:
ID:
713374
Start date:
27 May 2025
Project status:
Current
Principal investigator:
Ms Jianing Wang
Lead institution:
Huazhong University of Science and Technology, China

Research questions: Environmental factors play a crucial role in human health, yet their long-term effects on various health outcomes remain incompletely understood. Additionally, the underlying mechanisms and the interactions of environmental factors with genetic susceptibility and lifestyle factors require further exploration.
Objectives: To explore the associations between environmental exposures and multiple health outcomes, including neuropsychiatric diseases, brain disorders, and bone-related diseases. Additionally, this project will investigate the roles of genetic factors and lifestyle behaviors in these associations.
Scientific Rationale: Environmental factors play a crucial role in human health, yet their long-term effects on various health outcomes remain incompletely understood. Growing evidence suggests that air pollution, greenness, noise, and other environmental exposures contribute to the development of neuropsychiatric diseases (e.g., depression, anxiety), brain structure, and bone-related conditions (e.g., osteoporosis, fractures). However, the underlying mechanisms and the interactions of environmental factors with genetic susceptibility and lifestyle factors require further exploration. Exploring these questions is critical for disease prevention and public health interventions. This project aims to provide a systematic assessment of these relationships using the UK Biobank dataset.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-between-glycolipid-metabolism-and-major-adverse-cardiovascular-events-with-modifiable-lifestyle-factors-as-mediators-moderated-mediation-analyses-based-on-uk-biobank

Associations Between Glycolipid Metabolism and Major Adverse Cardiovascular Events with Modifiable Lifestyle Factors as Mediators: Moderated Mediation Analyses based on UK Biobank

Last updated:
ID:
91752
Start date:
21 December 2022
Project status:
Current
Principal investigator:
Professor Li Ling
Lead institution:
Sun Yat-Sen University, China

Cardiovascular disease (CVD) remains the most common cause of death globally and abnormal glycolipid metabolism greatly increases the risk of major adverse cardiovascular events (MACEs). People with abnormal glycolipid metabolism, especially those who have a diagnosis history of hyperlipidemia or diabetes, usually change their lifestyles according to the doctor’s advice, and therefore reduce the risk of MACEs. Therefore, modifiable lifestyle factors are likely to play a mediating role in the association between glycolipid metabolism and MACEs. Furthermore, the associations mentioned above might also vary in different strata or levels by demographic characteristics. Hence, we aim to evaluate the mediating role of modifiable lifestyle factors between glycolipid metabolism and MACEs and we hypothesize these associations are moderated by demographic characteristics. Findings from this research would provide valuable references for relevant healthy organizations to formulate healthy policies and guidelines as well as provide more practical, effective and personalized lifestyle advice for population with different demographic characteristics.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-between-healthy-lifestyle-socioeconomic-status-and-incident-mental-disorders-in-patients-with-type-2-diabetes-a-cohort-study

Associations between healthy lifestyle, socioeconomic status, and incident mental disorders in patients with type 2 diabetes: a cohort study

Last updated:
ID:
802140
Start date:
18 August 2025
Project status:
Current
Principal investigator:
Miss Guangcan Yan
Lead institution:
Harbin Medical University, China

Research questions: 1. To what extent do healthy lifestyle behaviors and socioeconomic status modify the risk of incident mental disorders in individuals with type 2 diabetes (T2D)?
2. Does socioeconomic status independently associate with mental health outcomes in T2D patients, beyond traditional clinical risk factors?
3. Are there synergistic or antagonistic interactions between healthy lifestyle and SES in mitigating mental disorder risks?
Objectives: This project aim to assess the associations of both healthy lifestyle and socioeconomic status (SES) with incident mental disorders in individuals with T2D. Additionally, we aim to examine weather overall lifestyles mediate associations of SES with incident mental disorders and the extent of interaction or joint relations of lifestyles and SES with health outcomes.
Scientific Rationale: T2D) is one of the most pressing challenges in global public health. Numerous studies have shown that the prevalence rate of mental disorders among individuals with T2DM is significantly higher than that in the general population. Understanding and evaluating the associated risk factors in this population can aid in developing effective intervention and prevention strategies to promote their mental health. Healthy lifestyle behaviors, as modifiable risk factors, along with SES, are closely associated with poor prognosis in T2DM. This study aims to assess these associations using exposure data (e.g., demographic and lifestyle factors) and health outcome data from the UK Biobank, thereby providing evidence for evidence-based practices.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-between-lifestyle-factors-overall-health-and-disease-risk-and-survival-in-the-uk-biobank

Associations between lifestyle factors, overall health and disease risk and survival in the UK Biobank

Last updated:
ID:
69371
Start date:
21 September 2021
Project status:
Current
Principal investigator:
Professor John C Mathers
Lead institution:
Newcastle University, Great Britain

Scientific rationale:
Lifestyle factors including body fatness, diet and exercise affect one’s risk of getting chronic diseases, such as cancer and cardiovascular disease, and may also be linked to chances of surviving after disease diagnosis. Associations between individual lifestyle factors, for example intake of dietary fibre, and the risk of, and survival from, diseases have been reported in other studies, as well as between scores and indices used to assess how healthy one’s lifestyle is. However, it important to understand the performance of these lifestyle scoring systems across different populations and, if required, to modify these to optimise their utility in future studies.

Aims:
This study aims to investigate relationships between such lifestyle factors, including the application of scoring systems used to assess lifestyle ‘healthfulness’, and overall health, risk of chronic diseases and survival. We also aim to explore how important each lifestyle factor is with respect to measuring lifestyle ‘healthfulness’ and whether we can improve the way in which we measure how healthy one’s lifestyle is (i.e. scoring systems).

How it will be done:
Over a three year project, we will use data on participant characteristics, lifestyle (e.g. diet, exercise, smoking and body fatness) from the UK Biobank Study. We will assess lifestyle ‘healthfulness’ for UK Biobank using, for example, existing scoring systems and indices. We will explore, using statistical techniques, whether individual lifestyle factors and measures of lifestyle ‘healthfulness’ predict the risk of developing diseases such as cancer and, in those who have been diagnosed with disease(s), their chances of survival.

Public Health Impact
The findings from this study could help to guide lifestyle recommendations and public health policies for the general population, and for those suffering from disease(s), to reduce the risk of chronic diseases and improve length, and quality, of life. By exploring the utility and quality of scoring systems and indices used to measure lifestyle ‘healthfulness’, this research could provide more accurate tools to be used in future studies worldwide.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-between-moderate-to-vigorous-physical-exercise-and-sedentary-behaviour-and-arterial-stiffness-and-blood-pressure-in-uk-adults

Associations between moderate to vigorous physical exercise and sedentary behaviour and arterial stiffness and blood pressure in UK adults

Last updated:
ID:
262
Start date:
1 December 2013
Project status:
Closed
Principal investigator:
Dr Soren Brage
Lead institution:
University of Cambridge, Great Britain

Physical activity (e.g. exercise or cycling to work) and sedentary behaviours (e.g. TV viewing) are both implicated in the development of heart disease and premature mortality. Intermediate heart disease markers include blood pressure and the stiffness of blood vessels.
The interplay of lifestyle exposures and their pathways to disease are not fully understood but are likely to involve both direct and indirect influence on the cardiovascular system. The role of adiposity as a potential mediator of these relationships remains uncertain, as does the degree to which these relationships are consistent across socio-demographic variables.
The aim of the current cross-sectional investigation is to examine the separate and combined associations between physical activity and sedentary behaviours with markers of vascular function in the UK adult population. This will inform prevention strategies that tackle important contributors to heart disease and early death in the UK.

This project involves a cross-sectional analysis of baseline variables. We require access to data only (not samples) for the full cohort.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-between-multimorbidity-and-breast-cancer-survival-a-uk-biobank-cohort-cross-sectional-study

Associations between multimorbidity and breast cancer survival: A UK Biobank Cohort cross-sectional study

Last updated:
ID:
95076
Start date:
16 February 2023
Project status:
Current
Principal investigator:
Miss Chidimma Maduabum
Lead institution:
Swansea University, Great Britain

Associations between multimorbidity and breast cancer survival: A UK Biobank Cohort cross-sectional study

Globally, breast cancer is the most typically diagnosed cancer among women. Age significantly impacts breast cancer, with older adults experiencing the most significant incidence rates. In the UK, between 2016 and 2018, on average, each year, 24% of new cases were in adults 75 and older. The occurrence of multiple chronic conditions in a person is referred to as multimorbidity. Comorbidity and multimorbidity are frequently used interchangeably. Compared to younger persons, multimorbidity is more common in older adults. People with common multimorbidity, especially older people who are most prone to developing cancer, are frequently excluded from clinical studies that guide cancer care. Therefore, it is necessary to understand how these specific diseases and their combinations impact breast cancer survival.
This research will investigate multimorbidity and breast cancer survival in the UK Biobank cohort. It will explore the association between multimorbidity and ethnicity as well as age in breast cancer survival.
This work aligns with the UK Biobank’s objective to advance the early detection, prevention, and treatment of significant illnesses, including breast cancer. This research will aid the proper identification and understanding of these associations between multimorbidity and breast cancer survival. The findings will improve breast cancer management strategies in routine clinical practice.
This study will use clinical data linkages and breast cancer outcomes data for the entire cohort to assess the pre-existing multimorbidity and their impact on breast cancer survival. The association between multimorbidity and breast cancer survival will be investigated using advanced statistical techniques and machine learning approaches.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-between-natural-and-built-environmental-factors-and-chronic-health-conditions-in-a-large-scale-cohort-in-the-united-kingdom

Associations between natural and built environmental factors and chronic health conditions in a large-scale cohort in the United Kingdom

Last updated:
ID:
73927
Start date:
6 July 2023
Project status:
Current
Principal investigator:
Dr Cui Guo
Lead institution:
University of Hong Kong, Hong Kong

Aim
This study aims to investigate the main and joint health effects of natural and built environmental factors on chronic health conditions in UK. The effect modifications by policy interventions and lifestyle will also be examined.

Scientific rationale
Both climate change and air pollution are the top leading risk factors for death and disease burden in the world. However, studies on the health effects of other natural and built environmental factors are limited and their potential pathways are inconclusive. This proposed study will depict the dose-response associations between natural/built environmental factors and chronic health conditions, and the potential thresholds might be observed. In addition, this study will identify the potential vulnerable population and laid a solid foundation for relevant targeted polices. The joint effects of natural and built environments will be further examined. Therefore, this project was proposed to expand previous studies by examining the health effects of comprehensive natural and built environmental factors and further exploring the potential vulnerable population.

Project duration
This project is expected to be completed within 36 months.

Public health impact
Due to rapid urbanization, the global burden of chronic diseases keeps increasing in past decades. This trend has brought substantial challenges to sustainable development. A few epidemiological studies have found closed associations between natural and built environmental factors and human health. However, the research findings remain insufficient and inconclusive. Hence, this proposed project will fill the gap in the diverse health effects of natural and built environmental factors and reveal their potential pathways. This project will also identify the vulnerable population, which provides scientific evidence for authorities to impose targeted polices and guidelines. The results findings will further unveil the interactions and joint health effects of natural and built environments. The research findings will provide robust and valuable evidence for urban managers to plan a strategic and healthy environment. In addition, this project will provide guidance for general population to develop a healthier lifestyle to promote their physical and mental health. Overall, this project will have significant implications for authorities to promote the improvement of public health, to develop healthy cities, and further to achieve the goal of sustainable development.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-between-neighbourhood-takeaway-food-environments-consumption-of-takeaway-food-and-body-weight

Associations between neighbourhood takeaway food environments, consumption of takeaway food and body weight.

Last updated:
ID:
9188
Start date:
1 February 2017
Project status:
Closed
Principal investigator:
Dr Thomas Burgoine
Lead institution:
University of Cambridge, Great Britain

Neighbourhood access to food outlets may play an important role in determining diet and health. This research will examine the extent to which neighbourhood takeaway food outlet exposure is associated with consumption of energy dense takeaway foods, body weight (body mass index and percent body fat) and likelihood of overweight and obesity in UK adults. Obesity is a risk factor for multiple chronic diseases, including type 2 diabetes, heart disease and stroke. Further, we will test for evidence of effect-modification of this relationship by sex, income, education, and genetic predisposition to obesity and disinhibited eating (developed from genotypic data). Mitigating the obesity epidemic is a UK public health priority. Governmental nutrition and health policies increasingly suggest that unhealthy neighbourhood takeaway food environments could be modified to promote healthier eating and improve health. However, the evidence-base to support such interventions remains weak. Through developing a better understanding of if and how food environments influence behaviour and health, this research will contribute to the UK obesity prevention strategy. As obesity is a risk factor for multiple chronic diseases, including type 2 diabetes, coronary heart disease and stroke, this research contributes to UK Biobank’s prevention of serious disease Aim. Using data for nearly 500,000 adults across England, I will use computerised mapping software (GIS) to capture exposure to takeaway food outlets in residential ‘neighbourhoods’ defined around individuals? home addresses. Locations of food outlets will be obtained from Local Authorities. I will then relate this exposure to consumption of takeaway food (from dietary intake data) and body weight (measured BMI and percent body fat) outcomes. We will explore the nature of these neighbourhood food environment effects across key individual, social and biological/genetic groups (derived from UK Biobank genotypic data) using statistical analyses testing for evidence of effect-modification. Full cohort.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-between-nr3c1-variants-and-metabolic-phenotypes-in-humans

Associations between NR3C1 variants and metabolic phenotypes in humans

Last updated:
ID:
65846
Start date:
11 January 2021
Project status:
Closed
Principal investigator:
Dr Mattia Quattrocelli
Lead institution:
Cincinnati Children's Hospital Medical Center, United States of America

Glucocorticoid steroids are widely used drugs to manage inflammation in many conditions. Chronic intake of these drugs promotes untoward side effects, like osteoporosis, obesity, growth suppression and hypertension. These steroids act through a receptor called glucocorticoid receptor, encoded by the gene NR3C1. This 3-year-long project aims at evaluating whether genetic variants in this gene correlate with alterations of metabolism or function. The results of this study may pave the way to better understand how we respond to these drugs and how we can counteract their side effects.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-between-parental-attained-ages-and-health-status

Associations between parental attained ages and health status

Last updated:
ID:
1417
Start date:
1 June 2013
Project status:
Closed
Principal investigator:
Professor David Melzer
Lead institution:
University of Exeter, Great Britain

Children of centenarians have lower prevalence of cardiovascular disease and live longer. Our recent work in the US Health and Retirement study (n=6500) showed large reductions in overall mortality in middle aged offspring for each decade their mothers or fathers lived beyond 65yrs. Estimates were little changed adjusting for classical risk factors. There was no effect of the parent’s attained age on spouse?s mortality. We showed parental survival associations with lower cancer incidence for the first time, but found no association with arthritis in offspring. We also found evidence of substantially lower rates of cognitive decline in offspring of long lived parents. These analyses suggest a strong intrinsic (probably genetic) influence explaining parent and offspring health advantage during ageing, and might provide a phenotype for understanding why some people suffer from age related disorders in their sixties while others remain disease free into their nineties and beyond.

We aim to estimate the associations between the full range of parental attained ages and health status (especially ageing traits) in UK Biobank respondents aged 55yrs and over. Given intergenerational gaps, the 55+ yr olds (estimated 308,000 participants) are more likely to have parents who have lived to very advanced ages. We aim to study parental longevity associations with common diseases in offspring, including diabetes, heart disease, stroke, common age related cancers and arthritis. We also want to estimate associations with common genetic variants (SNPs).

At present we plan to analyse the baseline (cross-sectional data) plus the data on all cause mortality already collected. We also request later access to data on incidence of our diseases of interest, deaths by cause, plus genetic variant (SNP) data, as these become available.

Our aim is to find new ways of delaying or treating age related disease and disability, to help people age well.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-between-physical-activity-patterns-and-measures-of-physical-function-in-south-asians-and-white-europeans

Associations between physical activity patterns and measures of physical function in South Asians and White Europeans

Last updated:
ID:
36371
Start date:
15 November 2018
Project status:
Closed
Principal investigator:
Professor Thomas Yates
Lead institution:
University of Leicester, Great Britain

South Asians (SAs) form the largest ethnic minority group in the UK. It has been established that SAs are at elevated risk of developing chronic diseases (e.g. diabetes/cardiovascular disease).
The aim of the proposed study is to investigate levels and prevalence of physical behaviours and function within SAs, whether these differ compared to white Europeans and whether observed differences are independent of sociodemographic factors. We will investigate whether physical behaviours/function and markers of metabolic health are associated with markers of cardiometabolic health in SAs and whether differences in these factors can help explain differences in health profiles between ethnicities. The proposed study is important as it will provide new quantified evidence comparing physical behaviours/patterns between SAs and WEs, the extent to which differences in these factors are independent or modified by sociodemographic factors and whether they contribute to the increased risk of chronic disease in SAs. Further understanding the role that physical behaviours/function have in contributing to the health status of SAs will help to better tailor and refine lifestyle interventions for this population and reduce the current health inequality seen across SA communities. This application will identify all those listed as SA within UK Biobank. We will then investigate levels and prevalence of physical behaviours/function in SAs and how this compares to the majority WE population. Regression analysis will examine the extent to identified differences in these factors are independent or modified by sociodemographic factors (e.g. education level, employment status, social deprivation) and whether they contribute to worse cardiometabolic health profiles within SAs. This analysis will aim to use the full cohort where possible. The cohort with accelerometer data will also be used.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-between-physical-multimorbidity-and-mental-health-outcomes-in-older-adults

Associations between physical multimorbidity and mental health outcomes in older adults

Last updated:
ID:
24776
Start date:
24 July 2017
Project status:
Closed
Principal investigator:
Dr Lucy Stirland
Lead institution:
University of Edinburgh, Great Britain

My research will explore links between having numerous physical illnesses and subsequent mental health problems, mainly in older people. It is known that physical ill-health often co-exists with poor mental health, but it is not fully understood which specific combinations of physical conditions are more detrimental to mental health. I will investigate various mental health problems that affect older people, including depression and dementia. In addition, I will examine blood pressure readings over time to evaluate if changes in blood pressure are linked with mental health. My aim is to improve our understanding of the links between physical multimorbidity and mental health with overall benefit to the general public. My findings will provide evidence on the interaction of chronic conditions which may guide their treatment, perhaps lending support to the idea of focusing less on individual diseases and more on the net consequences of complex multi-disease interactions. If my findings suggest that physical multimorbidity may contribute to mental ill health, then this may also be helpful in preventing mental illnesses including dementia by effectively managing upstream physical health determinants. I will start by examining all the data to look for overall links and patterns between having multiple physical illnesses and mental health problems. I will take into account other factors that are known to have an effect on both types of conditions, such as family history, socioeconomic status, smoking and alcohol intake. After this general exploration, I will look into specific combinations of diseases and their links with mental health. Finally, I also have a specific objective to look at the impact that changes in blood pressure across the life course have on mental health. I will request data from the full UK Biobank cohort.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-between-sleep-and-cognition-in-middle-aged-and-older-adults-mediating-effects-of-demographic-and-mental-health-factors-and-resting-state-brain-connectivity

Associations between sleep and cognition in middle-aged and older adults: mediating effects of demographic and mental health factors and resting state brain-connectivity

Last updated:
ID:
36328
Start date:
10 December 2018
Project status:
Closed
Principal investigator:
Dr Erika J Laukka
Lead institution:
Karolinska Institutet, Sweden

Whereas it is well-documented that sleep influences cognitive functions in young adults, studies on middle-aged and older adults have produced conflicting results. Age-related alterations in sleep duration and quality might help explain cognitive deficits with aging. On the other hand, there is also evidence that sleep-cognition associations diminish in older ages. We aim to examine the associations between different aspects of sleep and cognitive performance and determine when and for whom sleep has a strong impact on cognition and cognitive decline. In particular, we will investigate whether associations are modified by age, sex, or mental health status. Sleep may also affect brain efficiency, and thereby affect cognition. We will examine resting-state brain connectivity as a potential mediator in sleep-cognition association. The project will inform about the role of sleep for cognitive functioning in middle-aged and older adults and about the potential for improved sleep duration and quality as a possible intervention against age-related cognitive decline. Cross-sectional analyses will be performed on the whole cohort, including exploring the role of potential modifiers (age, sex, or mental health status). In subsequent parts of the project, the effect of sleep on cognitive decline will be explored for those who have follow-up data and resting-state brain connectivity as a possible mediator in the brain-cognition associations will be examined in the neuroimaging sample.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-between-the-24-hour-movement-behaviour-composition-and-dementia

Associations Between the 24-Hour Movement Behaviour Composition and Dementia

Last updated:
ID:
120034
Start date:
27 February 2024
Project status:
Current
Principal investigator:
Ms Margarita Liubetskaya
Lead institution:
Queen's University, Canada

This study aims to understand how an individual’s movement across the whole day, including the time they spend sleeping, sitting, and exercising, is related to their risk of developing dementia. Past research has looked at these behaviours one at a time. Our study aims to understand the collective effects of sleep, sitting, and exercise on dementia. The results of our research could help inform public health guidelines on how much sleep, sitting, and exercise are needed for good health. The project will take 1 year to complete.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-between-the-per3-vntr-polymorphism-and-sleep-circadian-cognitive-and-health-phenotypes-in-the-uk-biobank-wes-data

Associations between the PER3 VNTR polymorphism and sleep, circadian, cognitive and health phenotypes in the UK Biobank WES data

Last updated:
ID:
59619
Start date:
24 June 2020
Project status:
Current
Principal investigator:
Dr Simon Nicholas Archer
Lead institution:
University of Surrey, Great Britain

The circadian body clock regulates the daily temporal organisation of many of the body’s 24h rhythms including metabolism, immune function, cell growth, cognition, and the sleep-wake cycle. PERIOD3 (PER3) is one of the genes that regulates the circadian clock. In studies with small numbers of participants (up to 675), we have found that a particular genetic variant within PER3 is associated with diurnal preference, sleep-wake timing, brain activity, light sensitivity, cognitive performance, and a clinical condition called delayed sleep phase disorder where people go to bed and get up very late. Other small-scale studies have also shown links with bipolar disorder, diabetes, schizophrenia, cancer and addiction. The aim of this project is to investigate in greater detail the potential links between this PER3 variant with a much wider range of health conditions in the large UK Biobank cohort of participants.

The PER3 variant that we are interested in is a repeated section of DNA of up to 270 nucleotides (the individual building blocks of DNA). The UK Biobank holds a vast array of health-related data from 488,000 people and is also following the health of those people. It has also previously released genetic data from the same people but crucially those data only contain information about single nucleotide variants and not repeated sections like the one in PER3 that we are interested in. However, the UK Biobank has now released an updated database of genetic variants that does include repeated sections of DNA. So, the rationale for this project, which will last 3 years, is to investigate the PER3 repeat variant in a much larger group of people whose health status has been extensively characterised. This means that we will be able to determine in much more detail how this variant in a circadian clock gene affects short and long-term health. This particular PER3 variant is common in the UK population; around 10% of us have two copies of the rare variant and around 45% carry one copy. Therefore, a much greater understanding of how this PER3 variant contributes to health could have an important public health impact.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-air-pollutants-exposure-and-metabolic-and-genetic-changes-in-chronic-rhinosinusitis-and-allergic-rhinitis

Associations of air pollutants exposure and metabolic and genetic changes in chronic rhinosinusitis and allergic rhinitis

Last updated:
ID:
92566
Start date:
20 September 2022
Project status:
Current
Principal investigator:
Dr Ao Huang
Lead institution:
Huazhong University of Science and Technology, China

Aim: We hope to explore the effects of air pollutants exposure on the level of metabolism and gene expression in chronic rhinosinusitis (CRS) and allergic rhinitis (AR) patients by using the large-sample cohort omics data of UK Biobank

Scientific rationale: Ambient air pollution exposure remains a major public health threat worldwide and numerous studies have associated air pollution with increased risks of CRS and AR on an epidemiological level. Limited previous studies have demonstrated metabolic alterations and sequence variants in CRS and AR patients but upstream factors are not further explored. In studies on lower respiratory diseases such as asthma, exposure to air pollutants has been shown to affect peripheral blood metabolism and cause differential gene expression in patients. The evidence on relationship between ambient air pollution exposure and metabolism and genetic alterations in CRS and AR remains lacking. This study will help us to have a deeper understanding of the specific mechanism of occurrence and development in AR and CRS, and provide a new insight for future disease challenges.

Project duration: 36 months

Public health impact: Nasal inflammation diseases significantly affects people’s health, and air pollution is a major risk factor for these diseases. Exploring concrete mechanisms of air pollutants mediated the disease occurrence and development in multi-aspects can not only help us to more in-depth understanding of the disease but also can offer new evidence for global air pollution prevention and control, also help to make more rational health policy. At the same time, this research can also find new targets for disease intervention and provide a new direction for disease treatment.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-air-pollution-and-green-space-with-selected-chronic-diseases-and-their-interactions-with-genetic-susceptibility-and-lifestyle

Associations of air pollution and green space with selected chronic diseases and their interactions with genetic susceptibility and lifestyle

Last updated:
ID:
80741
Start date:
25 January 2022
Project status:
Current
Principal investigator:
Dr Haidong Kan
Lead institution:
Fudan University, China

While both air pollution and greenness have been associated with a wide range of health outcomes, more prospective cohort studies are warranted for causal pathways. Besides, current studies mainly target single disease-specific effect-response relationship, while cardiovascular disease and other non-communicable diseases (NCD) sharing potential pathways typically occurs in a context of multimorbidity. Whether individuals exposed to higher levels of environmental risk factors are more likely to develop multiple chronic conditions remains to be investigated. Furthermore, genome-wide association studies have identified potential genetic loci contributing to air pollution-related diseases. In addition, several aspects of lifestyle might affect health synergistically with environmental factors, serving as effect modifiers for exposure-outcome relationship. However, how genetic, lifestyle factors and environmental factors interact to cause or modulate diseases remains unclear.
This project aims to exaime associations between exposures to air pollution, greenness, noise, natural environment and selected chronic diseases, as well as potential interactions between those environmental factors and individual genotype and lifestyle factors (e.g., physical activity, dietary pattern, nutritional intake, sleep pattern) in this large-scale population study. Another interest is to examine the relationship between environmental exposures and multimorbidity. We will focus on cardiorespiratory health (such as atrial fibrillation, chronic obstructive pulmonary disease) and related traits (such as lung function), and NCD multimorbidity.
Specifically, we aim to 1) Assess the effect of various environmental exposures including air pollution, greenness, noise, natural environment on i) cardiovascular outcomes; ii) respiratory outcomes; iii) risk of NCD multimorbidity (the co-occurrence of chronic conditions); iv) other health outcomes of our interest; 2) Explore the potential interaction between these different environmental exposures; 3) Identify individuals with genetic susceptibility and the way health effect of environmental exposures can be modulated by individual genetic variation; 4) Examine whether lifestyle factors (e.g., physical activity, dietary pattern, nutritional supplementation, sleep pattern) play a role as potential effect modifier.
The project will be divided into several sub-projects focusing on several selected health outcomes (such as atrial fibrillation, chronic obstructive pulmonary disease, related traits, NCD multimorbidity) and is expected to last approximately 3 years. A better understanding of the interplay between different modifiable environmental factors and genetic predisposition is expected to facilitate more efficient prevention strategies and provide new evidence for urban planning framework. This project may lead to a more holistic approach to personalized prevention and treatment, accounting for both individual-level factors and community-level environmental factors.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-blood-biomarkers-with-cardiovascular-disease-and-related-cardiometabolic-outcomes-and-risk-prediction-in-the-clinical-setting

Associations of blood biomarkers with cardiovascular disease and related cardiometabolic outcomes and risk prediction in the clinical setting

Last updated:
ID:
9310
Start date:
1 June 2016
Project status:
Current
Principal investigator:
Professor Naveed Sattar
Lead institution:
University of Glasgow, Great Britain

In UK Biobank planned blood tests are important in helping detect early signs of groups of related diseases in the heart, blood vessels, brain, as well as early signs of diabetes. We will investigate to what extent these blood tests tell us about how likely someone is to develop these conditions, how these conditions develop, and whether we can intervene. For instance, adding information from these tests might improve our ability to predict the risk of a person having a heart attack. By harnessing the power of genes, we will test whether some of these new markers cause disease. This project will aim to assess avenues to improve health care throughout the population by investigating the improvement of CVD risk scores. More sensitive CVD and related risk scores may lead to better targeting of treatment and a reduction in the burden of CVD in the population. Biomarker measurement in UK biobank has been commenced, and the first tranche of biomarkers to be measured are now known. We will assess whether these markers are associated with, and predict, risk of cardiovascular and metabolic-related conditions. Biomarkers of interest include:
Lipids and lipoproteins (different measures of blood cholesterol), markers of inflammation, markers, of liver function, markers of renal function, sex hormones, markers of glucose control, and markers of bone health. Each of these has plausible biological mechanisms linking them to risk of cardiovascular and metabolic diseases. The full cohort with available data will be explored to maximise generalisability to the whole adult population.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-body-fat-and-inflammation-with-the-development-of-non-communicable-chronic-disease-a-prospective-cohort-study

Associations of body fat and inflammation with the development of non-communicable chronic disease: A prospective cohort study

Last updated:
ID:
144943
Start date:
13 March 2024
Project status:
Current
Principal investigator:
Dr Marcello Tonelli
Lead institution:
University of Calgary, Canada

Obesity associates with prevalent chronic disease, particularly type 2 diabetes, cardiovascular and pulmonary diseases, and cancers.

However obesity, defined as high body mass index (BMI), does not correlate with a higher risk of mortality in many clinical populations. In fact, when statistical modelling accounts for key confounders – inflammation and/or fasting insulin – high BMI (and high fat percentage) has been shown to associate with a lower risk of mortality in a general American adult population. These two confounders also associate with disease development. Neither confounder has been shown to be downstream from or caused by obesity. Given that adiposity functions as part of our immune system, obesity may arise as part of a protective response to disease development.
Besides BMI, there are many different ways and perhaps better ways to measure body fat (i.e., visceral fat, body adiposity index, body composition, relative fat mass, and waist circumference).

While there have been many systematic reviews noting the obesity paradox, (whereby mortality is lower for people with higher BMI) there have been few or no studies demonstrating that body fat, however measured, when accounting for inflammation, may protect against or delay the development of chronic disease.

In this prospective cohort study of UK adults, we will evaluate the associations of various body fat measures, independent of inflammation, with the development of chronic disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-cardiometabolic-genetic-and-lifestyle-factors-with-the-risk-of-arrhythmias

Associations of cardiometabolic, genetic, and lifestyle factors with the risk of arrhythmias

Last updated:
ID:
62306
Start date:
17 June 2020
Project status:
Closed
Principal investigator:
Mr Samuel Jie Tu
Lead institution:
University of Adelaide, Australia

Factors such as obesity, diet, and smoking play a significant role in the burden of cardiovascular disease globally. Identifying and managing factors that are modifiable is thus essential in the prevention and management of various cardiovascular diseases. While lifestyle-directed primary and secondary prevention strategies are well studied in heart attacks and heart failure, this is an emerging area in the field of heart rhythm disorders that requires further research.

We will use data obtained from the UK Biobank to identify associations between factors, particularly modifiable factors such as blood pressure, diet, physical activity, and weight, and the risk of developing heart rhythm disorders such as atrial fibrillation (irregular heart beats), bradyarrhythmias (slow heart beats) and sudden cardiac death. We will also investigate how the impact of these lifestyle factors may change with each individual’s unique genetic makeup.

Our work will build on the existing body of evidence and support current national and international guidelines for primary and secondary lifestyle interventions in the management of atrial fibrillation and other arrhythmias. Knowledge gained about how genetics and lifestyle factors interact with each other may serve to provide a precision medicine approach to the management of arrhythmias in the future.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-cardiovascular-and-lifestyle-factors-with-ocular-disease

Associations of cardiovascular and lifestyle factors with ocular disease

Last updated:
ID:
62103
Start date:
25 January 2021
Project status:
Current
Principal investigator:
Dr Michelle Sun
Lead institution:
University of Adelaide, Australia

There is an important relationship between heart and eye health. Various cardiovascular conditions such as high blood pressure, high cholesterol, heart disease and obesity have been previously shown to have a relationship with blinding eye conditions such as retinal artery and vein occlusion. There is also growing evidence that these heart conditions may have a role in the development and progression of glaucoma, the most common cause of irreversible blindness worldwide. It is still unclear which risk factors play the most important role, and how these eye conditions affect future risk of events such as heart attack and stroke. Our study will utilise the large UK Biobank database to better define these important relationships so that we can develop a more comprehensive understanding of the intricate relationship between the heart and the eye, leading to better detection, prevention and treatment of these common life and sight-threatening conditions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-chronic-diseases-with-behaviour-related-phenotypes-and-its-physiological-and-genetic-mechanisms

Associations of chronic diseases with behaviour-related phenotypes and its physiological and genetic mechanisms

Last updated:
ID:
781689
Start date:
14 July 2025
Project status:
Current
Principal investigator:
Professor Xueqiang Wang
Lead institution:
Wenzhou Medical University, China

Chronic diseases are highly prevalent in all populations, including the neuropsychiatric disorders, cardiometabolic multimorbidity and chronic pain. This study aims to investigate the associations between chronic diseases and behaviour-related phenotypes, and its physiological and genetic mechanisms. The behaviour-related phenotypes in this research primarily include the following aspects: lifestyle factors, sociodemographic indicators and physical measures. The physiological mechanism in this research primarily includes brain structure, blood cells and blood biochemistry, nuclear magnetic resonance (NMR) metabolomics, and proteomic phenotypes. Key research questions include: (1) How do behaviour-related phenotypes independently and interactively influence chronic disease risk? (2) What roles do brain structure, systemic inflammation, and metabolic dysregulation play in mediating these associations? (3) How do genetic predispositions and proteomics modulate these relationships across molecular and phenotypic levels?
The research will integrate multimodal data to address these questions. First, association between behaviour-related phenotypes and risk of chronic diseases were explored. Second, brain structure (regional grey matter volumes, white matter microstructure via MRI) will be analyzed to explore the links to chronic diseases and behaviour-related phenotypes. Inflammatory profiles (blood cell and biochemistry) and NMR metabolomics will assess systemic immunometabolic dysregulation. Genetic contributions will be evaluated through polygenic risk scores (PRS) for chronic diseases and traits, complemented by genome-wide association study (GWAS) to identify novel loci associated with disease and behaviour-related phenotypes interactions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-chronic-diseases-with-physical-activity-muscle-strength-lung-function-and-cognitive-function

Associations of chronic diseases with physical activity, muscle strength, lung function and cognitive function

Last updated:
ID:
92476
Start date:
25 August 2022
Project status:
Current
Principal investigator:
Professor Xueqiang Wang
Lead institution:
Shanghai University of Sport, China

The project will study the potential relationships between common chronic disorders (such as chronic low back pain, Parkinson’s disease, stroke and knee osteoarthritis), physical activity, muscle strength, and lung and cognitive functions. To explain further, physical activity means the time or intensity of daily activity, or other physical performances; muscle strength could be handgrip strength; lung functions indicate the healthy condition of the respiratory system while cognitive functions mean the variety of mental processes, including memory and decision making. Based on previous studies, the morbidity of cardiometabolic multimorbidity and dementia is associated with handgrip strength, and the risk of incident arrhythmias is related to physical activity. Therefore, it is rational to hypothesise that time or intensity of daily activity, handgrip strength, lung health, or memory condition is related to the happening or development of these common chronic disorders.
The duration of this project is approximately 3 years. The results of this project will indicate the potential associations between chronic diseases and other functions, such as intensity of daily activity, handgrip strength, healthy lung conditions and memory. Therefore, these findings could tell people about possible causes or factors of the development of stroke, and knee osteoarthritis, thereby providing more information for clinicians and therapists to prevent the happening and decrease the risk of death from these disorders.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-circulating-25-hydroxyvitamin-d-concentrations-with-colorectal-cancer-risk-and-survival

Associations of circulating 25-hydroxyvitamin D concentrations with colorectal cancer risk and survival

Last updated:
ID:
69359
Start date:
25 January 2021
Project status:
Current
Principal investigator:
Dr Dong Hang
Lead institution:
Nanjing Medical University, China

Colorectal cancer is the third most common cancer and the second leading cause of cancer death globally. The etiology and prognosis of colorectal cancer are closely related to dietary and nutritional factors, among which vitamin D has been a topic of considerable interest. However, population-based evidence remains inconclusive regarding the associations between vitamin D status and colorectal cancer risk and survival. For risk analysis, the inconsistency is probably due to the differences in sample sizes, assays, and laboratories. Moreover, there are still very limited data on the exact dose-response association between 25(OH)D and colorectal cancer risk. For survival analysis, the observation of considerable between-study heterogeneity (e.g., populations, assay methods, and definitions of vitamin D categories) limited the validity of summary estimates. Furthermore, the different time of blood draw (including pre-diagnostic, post-diagnostic, pre-operative, and post-operative) for vitamin D measurements may confuse the results, and pre-diagnostic vitamin D status seems more stable and useful to assess the proposed prognostic value for colorectal cancer patients. Therefore, leveraging the UK Biobank resource, a large cohort with a long-term follow up, we aim to prospectively evaluate the associations of season-standardized 25(OH)D concentrations with CRC risk. We anticipate that this project will take up to 12 months. The results of this project will improve our understanding about the associations of vitamin D status with colorectal cancer risk and survival, and help to develop new strategies for colorectal cancer prevention and treatment.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-combined-genetic-and-lifestyle-risks-with-incident-cardiovascular-disease-and-cancers

Associations of Combined Genetic and Lifestyle Risks With Incident Cardiovascular Disease and Cancers

Last updated:
ID:
92014
Start date:
20 October 2022
Project status:
Current
Principal investigator:
Dr Shaoyong Xu
Lead institution:
Hubei University of Arts and Sciences, China

Cardiovascular disease and cancers are the leading cause of mortality and morbidity worldwide and is driven by both genetic and lifestyle factors. Recent cohort studies have indicated that adherence to a healthy lifestyle plays a pivotal role in attenuating the impact of genetic factors on risk of several chronic diseases. Lifestyle factors are commonly viewed as mediators between genetic factors and risk of several chronic diseases and that healthy lifestyles might alleviate the disease incident in different genetic risk. Multiple studies have examined the contribution of an individual lifestyle factor or several in the association between genetic risk and mortality or morbidity of cardiovascular disease and cancer. However, important gaps remain. First, how much an overall lifestyle mediates the association between genetic and health outcomes is debatable. Besides, lifestyle factors are interrelated and few studies have built a healthy lifestyle score to reflect overall lifestyle and to evaluate its impact on the genetic risk in health. Second, limited research has been performed on the interaction and joint associations of genetic and overall lifestyles with health outcomes. Third, it remains unclear whether the findings are consistent among sub-populations of different age, sex, and racial or ethnic groups. In this study, we will examine whether overall lifestyles mediate associations of genetic with incident cardiovascular disease and cancers and the extent of interaction or joint relations of lifestyles and genetic with health outcomes. The project duration is about 3 years. Clarifying the associations of combined genetic and lifestyle risks with incident cardiovascular disease and cancers will help guide population prevention strategies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-diet-and-metabolic-profile-with-risk-of-cardiometabolic-disease-and-neurodegenerative-disease

Associations of diet and metabolic profile with risk of cardiometabolic disease and neurodegenerative disease

Last updated:
ID:
96083
Start date:
15 November 2022
Project status:
Current
Principal investigator:
Dr Xiang Gao
Lead institution:
Fudan University, China

Aims: To investigate the associations of different dietary patterns with risk of cardiometabolic diseases (cardiovascular disease [CVD], type 2 diabetes [T2D], chronic kidney disease [CKD]) and neurodegenerative disease (Alzheimer’s disease and Parkinson’s disease). In addition, to test whether the associations could be modified/mediated through the circulating multi-omics level outcomes. Moreover, to investigate whether adding the selected multi-omics level outcomes could improve the prediction of the development of cardiometabolic diseases and neurodegenerative disease.

Rationale: Cardiometabolic disease is a serious public health crisis including a group of chronic diseases including coronary heart disease, stroke, diabetes, and CKD. In addition, neurodegenerative disease, e.g. Alzheimer’s disease and Parkinson’s disease is the leading cause for disability of the older adults. Diet is one of the most important contributors and modifiable risk factors of cardiometabolic diseases and neurodegenerative disease. Dietary pattern approach that considers the complexity of diet has emerged as a useful tool, represent a broader picture of food and nutrient consumption, and may thus be more predictive of disease risk than individual foods or nutrients. Although growing evidence has linked different dietary patterns with risk of cardiometabolic diseases, the association of dietary patterns with risk of neurodegenerative disease is relatively under-investigated. Furthermore, whether the associations of dietary patterns with cardiometabolic diseases and neurodegenerative disease could be modified or mediated by multi-omics level outcomes have not been well scrutinized, which are critical for understanding the causes of disease and making strategies for prevention/treatment.

In addition, nuclear magnetic resonance (NMR) emerging as a promising tool for detecting metabolites and providing absolute quantitation with relatively lower costs and better reproducibility, has been increasingly applied to assess the associations of biomarkers and various of common diseases such as CVD and T2D. However, whether adding the selected multi-omics level outcomes could improve the prediction of the cardiometabolic diseases and neurodegenerative disease is limited.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-dietary-exposome-with-inner-retinal-thickness-parameters-in-the-uk-biobank

Associations of dietary exposome with inner retinal thickness parameters in the UK Biobank

Last updated:
ID:
151366
Start date:
7 March 2024
Project status:
Current
Principal investigator:
Dr Bénédicte MJ Merle
Lead institution:
University of Bordeaux, France

Glaucoma is the second leading cause of blindness worldwide and its prevalence is projected to reach 111.8 million in 2040. This progressive degenerative optic neuropathy is characterized by the irreversible and progressive degeneration of retinal nerve cells. At the beginning peripheral vision is lost and if the lesions progress, central vision disappears, leading to blindness. Visual impairment due to glaucoma has major consequences on quality of life and loss of autonomy, with considerable personal, familial and societal burden. Hence, a preventive approach, aiming at reducing the impact of risk factors in order to delay the disease’s onset is of utmost importance.
In addition to existing treatments for glaucoma, preserving the nerve functions of the eye is of utmost importance. A high-quality ad healthy diet provides nutrients with neuroprotective and anti-inflammatory properties. Those nutrients are useful for maintaining good eye health. Little is known about the impact of our diet on glaucoma.
The aim of this project is to characterize the associations between our diet and the loss of retinal nerve cells, a well-established glaucoma biomarker.
We will use the unique data of the UK Biobank where the loss of retinal nerve cells was measured using optical coherence tomography (OCT), i.e. eye scan. UK Biobank participants also filled extensive dietary questionnaires allowing the computation of dietary scores and patterns such as the well know Mediterranean diet. Dedicated statistical model will be performed to evaluated the link between retinal thickness and dietary exposure in about 65 900 participants.
This project will improve the understanding of the impact of our dietary consumption in glaucoma. This could open new perspectives of preventive strategies to delay or avoid the occurrence of glaucoma.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-dietary-factors-and-vitamin-supplement-use-with-risk-of-covid-19-chronic-disease-and-mortality

Associations of Dietary Factors and Vitamin Supplement Use with Risk of COVID-19, Chronic Disease and Mortality

Last updated:
ID:
66163
Start date:
22 September 2020
Project status:
Current
Principal investigator:
Dr Demetrius Albanes
Lead institution:
National Cancer Institute, United States of America

Long-standing interest has been put on health effects of nutritional factors through biological mechanisms involved in pathways including ROS, inflammation, hormones/energy metabolism, gene expression regulation, immunity, and cell cycle/proliferation. Nutritional factors played important roles in noncommunicable diseases including diabetes, cancer and cardiovascular diseases, which accounted for almost 70% of all deaths worldwide. In addition to the role of individual food items, analysis of dietary patterns has gained increasing importance because specific nutrients are consumed across a wide range of foods and complex food components can have competing biological effects. Therefore, integrated analyses of dietary patterns that reflect more diverse and multi-dimensional nutritional compositions are needed to relate to health outcomes and facilitate translational dietary recommendations. Additionally, interactions between genetic variation in nutritional metabolism and dietary factors have been shown for risk of chronic diseases that deserve greater attention. Beyond above-mentioned beneficial chronic disease health associations, it is of timely importance to determine whether these nutritional factors are related to risk of COVID-19 and have any preventative potential against infection. The role of diet and vitamin and micronutrient supplements has been the focus of chronic disease etiologic and prevention research for decades through prospective cohorts, nested case-control biochemical studies, and controlled trials. In the field of cancer for example, there has been interest in whether and how vitamin D impacts risk and survival, and most recently interest in vitamin D for prevention of respiratory infections including COVID-19 has re-emerged. In addition, how genetic variation modifies nutritional and biochemical metabolism and influences vitamin supplementation effects on chronic disease and risk of COVID-19 and prognosis remain unknown.

The overarching aim of this proposal is to comprehensively explore associations of dietary factors with risk of COVID-19, chronic diseases and mortality. Specifically, we aim to: 1) Examine associations of Healthy Eating Patterns, single nutritional factors and dietary supplement use with risk of COVID-19, cancer, diabetes and mortality; 2) Assess whether genetic variants associated with nutritional and biochemical metabolism can modify the observed associations. 3) Study whether key lifestyle risk factors and racial/ethnic groups can modify the observed associations. Project duration will be 36 months. Our findings may provide evidence that dietary factors and possible modification may impact long-term health outcomes as well as risk of and prognosis for COVID-19 infection. The proposal should also facilitate development of general dietary guidelines, promote translational findings into dietary recommendations and contribute to ongoing informed public health practices.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-early-life-factors-genes-adulthood-factors-and-diseases

Associations of early-life factors, genes, adulthood factors, and diseases

Last updated:
ID:
96054
Start date:
4 April 2023
Project status:
Current
Principal investigator:
Professor Shunqing Xu
Lead institution:
Hainan University, China

The rising prevalence of adulthood diseases such as type-2 diabetes and obesity has been recognized as a public health problem globally. In addition to lifestyle, environmental factors in adulthood, and genetic variations, developmental conditions in early life also have essential roles in adult health and disease. However, studies considering multi factors, such as early-life, genetic, life-long weight/body-size changes, and adulthood risk factors, simultaneously in predicting diseases are still lacking. Therefore, we intend to investigate and quantify the associations of early-life factors, genes, adulthood factors, the combination of these factors, and diseases using the UK Biobank database.

This project will last for 36 months.

Findings from this project will facilitate targeted preventive strategies for adult diseases with cost-effectiveness by modifying factors closely associated with the risk factors contributing the most to diseases, such as early life, genetic, or adulthood factors.
We propose to use the whole UK Biobank cohort in our analysis.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-environmental-and-genetic-factors-with-the-risks-of-incidence-and-progression-of-cardiometabolic-diseases

Associations of environmental and genetic factors with the risks of incidence and progression of cardiometabolic diseases

Last updated:
ID:
109546
Start date:
18 October 2023
Project status:
Current
Principal investigator:
Professor Kai Huang
Lead institution:
Union Hospital, Tongji Medical College, China

The overall objective of the research is to improve the quality of life for the patients with cardiovascular related diseases.The research includes two parts’ data, analysing our own cohort data and UK biobank data. The main purpose of this study is to verify that environmental exposure (lifestyle, diets, psychological factor, economic conditions, etc.), and genetic factors which have significant effects on these disease’s development or regression. In addition, the investigators will further study the genetic mechanism of these diseases from the molecular genetic level, and explore the contribution and impact of genetic diversity on the treatment effect. At the same time, explore and establish a cardiovascular related diseases prevention and control system and effective mechanism of hospital.This research will provide theoretical and experimental basis for formulating strategies for large-scale intervention of related diseases, so as to improve the prevention, treatment and prognosis. We strive to reduce the disease and social burden.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-environmental-and-metabolic-risk-factors-and-the-interactions-with-genetic-susceptibility-loci-with-risk-of-covid-19-chronic-disease-and-mortality

Associations of environmental and metabolic risk factors, and the interactions with genetic susceptibility loci with Risk of COVID-19, Chronic Disease and Mortality

Last updated:
ID:
98583
Start date:
1 March 2023
Project status:
Current
Principal investigator:
Professor Jiaqi Huang
Lead institution:
Second Xiangya Hospital of Central South University, China

Long-standing interest has been put on the health effects of environmental and metabolic risk factors through biological mechanisms involved in metabolic pathways including ROS, hormones, inflammation, energy metabolism, gene expression regulation, immunity, and cell cycle/proliferation. Environmental and metabolic risk factors have key roles in diseases including diabetes, cancer and cardiovascular diseases, which may account for over 80% of all deaths worldwide. In addition to investigate individual factors, investigations of combined risk factors on disease and mortality risk have gained substantial attention given their synergistic or antagonistic effects. Therefore, integrated analyses of dietary behaviors, lifestyle, social and natural environment, physical examination and laboratory biomarkers, which represent more diverse and multi-dimensional exposures, are needed to investigate potential etiologies of chronic diseases and facilitate translational health-care policies. Moreover, interactions between genetic variation and above risk factors have been shown for risk of chronic diseases including cancer and mortality that may shed light on the biological mechanisms and deserve greater attention. Beyond the above-mentioned beneficial chronic disease health associations, it is of timely importance to determine whether these factors are associated with the risk of COVID-19 and death, and have any potential preventative potential against infection and progression.

The overall aim of this proposal is to comprehensively examine associations of environmental and metabolic risk factors and the interactions with genetic susceptibility loci with risk of COVID-19, chronic diseases and mortality. Specifically, we aim to: 1) Assess associations of dietary behaviors, lifestyle, natural and social environment, physical examination and laboratory biomarkers with risk of COVID-19, cancer, diabetes, and overall and cause-specific mortality; 2) Evaluate whether genetic variants can modify the associations of dietary behaviors, lifestyle, natural and social environment, physical examination and laboratory biomarkers with risk of COVID-19, chronic diseases and mortality; 3) Explore whether age, gender, racial/ethnic group, genetic background, and other selected factors can modify the associations of dietary behaviors, lifestyle, natural and social environment, physical examination and laboratory biomarkers with risk of COVID-19, chronic diseases and mortality. Project duration will be 36 months and it might be extended given advances in statistical methodology and additional validation of findings. Our findings may provide evidence that environmental and metabolic factors and possible modification may impact long-term health outcomes as well as risk and prognosis for COVID-19 infection. The project should also facilitate development of standards of medical care, promote translational findings into health-care policies and contribute to ongoing informed public health practices.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-genetic-factors-healthy-lifestyle-behavior-and-environmental-pollution-with-chronic-respiratory-diseases

Associations of genetic factors, healthy lifestyle behavior, and environmental pollution with chronic respiratory diseases

Last updated:
ID:
76194
Start date:
26 August 2021
Project status:
Current
Principal investigator:
Professor Chunhui Ni
Lead institution:
Nanjing Medical University, China

Background: Chronic respiratory diseases have become a public health problem worldwide. The development of chronic respiratory diseases is a complex interaction between genetic risk and environmental factors. Environmental risk factors (i.e. PM2.5), unhealthy lifestyle factors (i.e. smoking) and genetic risk (i.e. polygenic risk score) has been widely associated with the risk of chronic respiratory diseases. However, previous studies usually ignored the interactions between these environmental factors and genetic risk.

Objective: In this study, we mainly want to comprehensively explore the interactions between genetic risk (polygenic risk scores) and environmental risk factors (including unhealthy lifestyle factors) for overall chronic respiratory diseases.

Scientific Principles: This study aims to comprehensively investigate the associations of genetic factors, common environmental factors, and lifestyle factors with of chronic respiratory diseases risk and explore the interactions between genetic and environmental factors. Then, we include the interaction terms as well as genetic and environmental factors to build a risk prediction model, and examine the improvement of risk prediction.

Project duration: Data analysis and the publication of the findings will be completed within 36 months.

Public health impact: This project is expected to identify interactions between genetic risk and environmental factors for of chronic respiratory diseases, and improve the efficiency of risk prediction. The results will help identify groups of individuals who are at higher risk of chronic respiratory diseases and guide early, targeted prevention by combining genetic and environmental factors.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-genetic-factors-healthy-lifestyle-behavior-and-environmental-pollution-with-common-chronic-diseases

Associations of genetic factors, healthy lifestyle behavior, and environmental pollution with common chronic diseases

Last updated:
ID:
617693
Start date:
23 July 2025
Project status:
Current
Principal investigator:
Mrs Ting Wang
Lead institution:
Nanjing Drum Tower Hospital, China

This project aims to investigate the associations of genetic factors, healthy lifestyle behaviors, and environmental pollution with the development of common chronic diseases, such as cardiovascular diseases, diabetes, and respiratory disorders. Using data from the UK Biobank (UKB), the research will address the following key questions:

1. How do genetic risk interact with modifiable lifestyle factors (e.g., diet, physical activity, smoking) to influence the risk of chronic diseases?

2. What is the role of environmental pollution (e.g., air pollution, noise) in modifying these associations?

3. Can a healthy lifestyle mitigate the adverse effects of genetic risk and environmental exposures on chronic disease outcomes?

The objectives are:

1. To identify genetic variants associated with chronic diseases and assess their interaction with lifestyle and environmental factors.

2. To quantify the independent and joint effects of genetic risk, lifestyle behaviors, and environmental pollution on disease risk.

3. To explore potential mechanisms underlying these interactions using multi-omics data (e.g., genomics, metabolomics).

The scientific rationale is rooted in the growing burden of chronic diseases globally, which are influenced by a complex interplay of genetic, behavioral, and environmental factors. Understanding these interactions can inform personalized prevention strategies and public health interventions to reduce disease risk, particularly in populations with high genetic susceptibility or environmental exposures. The UKB’s rich phenotypic, genetic, and environmental data provides a unique opportunity to address these questions comprehensively.

This research will contribute to the development of targeted interventions that integrate genetic risk profiling, lifestyle modification, and environmental policy to improve population health outcomes.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-genetic-factors-healthy-lifestyle-behavior-and-environmental-pollution-with-esophageal-diseases

Associations of genetic factors, healthy lifestyle behavior, and environmental pollution with esophageal diseases

Last updated:
ID:
90820
Start date:
9 August 2022
Project status:
Current
Principal investigator:
Dr Jing Xu
Lead institution:
Nanjing Medical University, China

Background: The incidence of esophageal diseases such as esophageal adenocarcinoma (EAC) and gastroesophageal reflux disease (GERD) have experienced an evident increase over the past decades and become a public health problem worldwide. And the incidence of EAC has become more prevalent while the incidence of most other solid tumors has seen a decrease over the last decades. The development of esophageal diseases is usually a complex interaction between genetic risk and environmental factors. Environmental risk factors, unhealthy lifestyle factors, and genetic risk (i.e. polygenic risk score) has been widely associated with the risk of esophageal diseases.

Objective: In this study, we mainly want to comprehensively explore the interactions between genetic risk (polygenic risk scores) and environmental risk factors (including unhealthy lifestyle factors) for overall esophageal diseases.

Scientific Principles: This study will firstly investigate the associations of genetic variants, environmental factors, and lifestyles with esophageal disease risk. Then, this study will also explore the interactions between genetic and environmental factors. At last, we plan to build a risk prediction model for esophageal disease with the identified risk factors.

Project duration: Data analysis and the publication of the findings will be completed within 36 months.

Public health impact: This project is expected to identify risk factors for esophageal diseases, and help identify individuals who are at higher risk of esophageal diseases to guide early prevention.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-genetic-factors-healthy-lifestyle-behavior-testosterone-level-and-sleep-traits-with-human-healthspan

Associations of genetic factors, healthy lifestyle behavior, testosterone level and sleep traits with human healthspan

Last updated:
ID:
64689
Start date:
14 July 2021
Project status:
Current
Principal investigator:
Dr Juncheng Dai
Lead institution:
Nanjing Medical University, China

Life expectancy has increased dramatically since the last century, while the quality of life for the elderly has not increased proportionally. These challenges call for longevity research to focus on understanding the pathways controlling healthspan.
Previous studies have found that endogenous testosterone concentrations, lifestyle behaviors, and sleep traits are associated with the risk of various cancers and chronic diseases, while the relationship between them and healthspan is still unknown.
Complex mechanisms driven by a combination of genetic, environmental, and lifestyle factors cause differences in individual healthspan. UK Biobank cohort, a large dataset with both environmental exposures and genomics data, has an incomparable advantage to systematically identify the risk factors for short healthspan.
We estimate that we will complete the majority of data analysis and finish the publication of the relevant results in 18 months. For these aims, we propose to:
1. investigate the associations and the causal relationships between testosterone level, sleep traits, healthy lifestyle index (HLI), and healthspan;
2. develop a polygenic risk score (PRS) for human healthspan based on reported healthspan-associated genetic variants;
3. use the above results to generate a health prediction model to establish new strategies for extending healthspan.
Our study will help to promote the understanding of aging and longevity, and the model could be used in identifying groups of individuals who are at high risk of short healthspan and more likely to benefit from interventions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-genetic-socioeconomic-status-lifestyle-and-environmental-factors-with-health-outcomes

Associations of genetic, socioeconomic status, lifestyle, and environmental factors with health outcomes

Last updated:
ID:
91019
Start date:
4 May 2023
Project status:
Current
Principal investigator:
Professor Chun-Quan Ou
Lead institution:
Southern Medical University, China

Aims: This project aims to (1) investigate the associations between genetic, socioeconomic status, (time-varying) lifestyle, environmental factors and health outcomes; (2) further determine potential routes among socioeconomic status, various exposures and health outcomes; (3) reveal the influence of covariate missing on estimates of associations and compare different imputation methods.

Scientific rationale: As the population is aging, non-communicable diseases such as cardio-respiratory, metabolic, and neurodegeneration diseases are increasingly prevalent in these years. Previous studies have identified several determinants of the occurrence and mortality of diseases of middle- and old-aged individuals, including genetic factors, cigarette smoking, alcohol consumption, air pollution, diet, and physical activity, etc. Assessing the independent and joint effects of genetic factors, socioeconomic status, (time-varying) lifestyle, environmental factors on health outcomes and further elucidating the underlying mechanisms using statistical approaches would shed light on targeted interventions and promote healthy aging.

Project duration: 36 months

Public health impact: The findings of this project would have important implications on how to promote healthy aging and subsequently improve public health through targeted interventions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-genetics-lifestyles-and-environmental-risk-factors-with-age-related-diseases-and-aging

Associations of genetics, lifestyles, and environmental risk factors with age-related diseases and aging

Last updated:
ID:
362917
Start date:
28 October 2024
Project status:
Current
Principal investigator:
Professor Youxin Wang
Lead institution:
North China University of Science and Technology, China

This research project aims to investigate the effect of genetics, lifestyles (diet, smoking, and physical activities, etc.), and environmental (non-genetic/modifiable) risk factors on age-related diseases (hypertension, diabetes, cancers, Alzheimer’s disease, Parkinson’s disease, osteoporosis, kidney failure, chronic obstructive pulmonary disease, multiple sclerosis, amyotrophic lateral sclerosis, cognitive function, and mental health, etc.), mortality, life expectancy, and aging. Furthermore, this project also aims to explore the interaction and joint associations of genetics, lifestyles, and environmental determinants with age-related outcomes.

The aging of the global population is accelerating due to longer life expectancy. Age-related diseases refer to medical conditions that occur with advancing age, including hypertension, diabetes, cancers, Alzheimer’s disease, Parkinson’s disease, osteoporosis, kidney failure, chronic obstructive pulmonary disease, multiple sclerosis, and amyotrophic lateral sclerosis. These diseases remain the leading causes of death in the global elderly population. The administration of age-related diseases requires advanced medical services and long-term healthcare support. Studies indicate that extending a healthy lifespan by one year holds the possibility to decrease approximately £2,000 annually in health-related costs. The development of age-related outcomes is determined by numerous factors, including but not limited to genetics, lifestyles, and environmental risk factors. Healthier lifestyles, including stopping smoking, increasing physical activity, and maintaining a balanced diet, have been widely recognized as crucial factors in alleviating the impact of age-related disease. In light of the growing availability of genetic research, accumulating evidence highlights the contribution of genetics to the variation in age-related diseases and aging. Research has also shown that the genetic and environmental risk factors can contribute to age-related diseases and influence the aging process. However, the exact impact of these factors and the biological mechanisms underlying the progression of these outcomes remain unexplored. Collectively, it is indispensable to understand the interaction and joint associations of genetics, lifestyles, and environmental risk factors with age-related diseases, mortality, life expectancy, and aging.

The study is expected to last approximately 36 months.

Current research results would be utilized to optimize healthcare resource allocation, formulate precise public health strategies, and reduce the burden of age-related diseases and aging.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-genomic-environmental-clinical-information-and-biomarker-with-chronic-non-communicable-diseases

Associations of genomic, environmental, clinical information, and biomarker with chronic non-communicable diseases

Last updated:
ID:
104277
Start date:
5 October 2023
Project status:
Current
Principal investigator:
Professor Ming Lu
Lead institution:
Shandong University, China

Chronic non-communicable diseases, such as cancer, cardiovascular and cerebrovascular diseases, chronic respiratory diseases, mental and nervous system diseases, etc., have caused a considerable burden worldwide. Previous studies have reported some potential risk factors for chronic diseases. However, how other factors and their interactions impact the development of chronic diseases remains to be explored. UK Biobank is a large prospective natural population cohort containing high quality individual data and extensive information. We propose to leverage the UK Biobank to explore the individual and combined effects of genetics, socioeconomic status, lifestyle, environmental exposure, clinical features on chronic noncommunicable diseases. In addition, risk prediction biomarkers for specific chronic noncommunicable diseases will be identified. The duration of this project is expected to last 36 months.
This study will help clarify the relationship between various risk factors and chronic non-communicable diseases and identify high-risk individuals, which has important public health implications for the precise prevention and management of chronic non-communicable diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-gut-microbiome-related-exposures-with-early-onset-colorectal-cancer

Associations of gut microbiome-related exposures with early-onset colorectal cancer.

Last updated:
ID:
538807
Start date:
24 March 2025
Project status:
Current
Principal investigator:
Dr Chengzhi Huang
Lead institution:
Guangdong Provincial People's Hospital, China

Early onset colorectal cancer is becoming increasingly common, making it crucial to understand its causes. The gut microbiome is likely involved in the development of key factors that contribute to this type of cancer. Are there specific gut microbiome biomarkers that can be used for the early diagnosis and screening of early-onset CRC? My research plan is to use data from the database to explore the role of gut microbiome on the development and progression of colorectal cancer and identified the specific intestinal flora that had a causal relationship with the risk and prognosis of colorectal at the level of gene prediction, which may provide helpful biomarkers for early disease diagnosis and potential therapeutic targets.
Dysbiosis of the gut microbiome is a hallmark of colorectal cancer development. As precancerous lesions like adenomas progress to colorectal cancer, a decline in microbial diversity and an increase in pathogenic bacteria associated with cancer have been observed. These pathogenic bacteria contribute to tumor initiation and progression through several mechanisms: they produce carcinogenic genotoxins, interact with host cell receptors via bacterial adhesins, metabolize dietary components to generate tumor metabolites, and engage with genetic or epigenetic changes.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-habitual-sleep-duration-and-healthy-diet-with-digestive-system-cancer-mortality-in-the-uk-biobank-cohort

Associations of Habitual Sleep Duration and Healthy Diet with Digestive System Cancer Mortality in the UK Biobank Cohort

Last updated:
ID:
694569
Start date:
16 June 2025
Project status:
Current
Principal investigator:
Dr Jingyuan Zhang
Lead institution:
Hubei University of Medicine, China

Research Questions:
What is the association between habitual sleep duration and mortality due to digestive system cancers in the UK Biobank cohort?
How does adherence to a healthy diet modify the risk of digestive system cancer mortality?
Are the associations between sleep duration, dietary habits, and digestive system cancer mortality influenced by factors such as smoking, body mass index (BMI), or type 2 diabetes?
Objectives:
To evaluate the relationship between short (!6 hours), normal (7-8 hours), and long (!9 hours) sleep durations and the risk of digestive system cancer mortality.
To investigate the protective effects of a healthy diet-characterized by higher fiber intake, greater consumption of fruits and vegetables, and limited meat consumption-on digestive system cancer mortality.
To explore the potential interactions and mediating effects of lifestyle factors (e.g., smoking, BMI, type 2 diabetes) on the associations between sleep duration, diet, and cancer mortality.
Scientific Rationale:
Digestive system cancers are among the leading causes of cancer-related mortality worldwide. Lifestyle factors such as diet and sleep are modifiable risk factors that may influence the development and progression of these cancers. While previous studies have independently examined the impacts of sleep and diet, few have explored their combined effects on digestive system cancer mortality. This research leverages the UK Biobank cohort, a large-scale dataset with detailed lifestyle and health information, to provide novel insights into the interplay between sleep, diet, and cancer mortality. Understanding these relationships can inform public health strategies to reduce cancer burden and improve survivorship.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-healthy-lifestyle-with-incident-major-non-communicable-chronic-diseasesncds-and-mortality

Associations of Healthy Lifestyle with Incident Major Non-communicable Chronic Diseases(NCDs) and Mortality

Last updated:
ID:
482700
Start date:
16 December 2024
Project status:
Current
Principal investigator:
Dr Xiang Gao
Lead institution:
Shanghai Jiao Tong University School of Medicine., China

Aims: This research aims to understand how various lifestyle factors such as diet, exercise, smoking, sleep, and social connections influence the risk of developing major non-communicable diseases (NCDs) like heart disease, diabetes, and cancer, as well as the risk of dying from any cause. We seek to determine what proportion of these risks could be reduced by making healthier lifestyle choices.
Scientific Rationale: Despite advancements in medical technology and healthcare, NCDs remain the leading cause of death globally, largely due to modifiable lifestyle factors. There is a great need for a theoretical framework based on lifestyle medicine that systematically assesses lifestyle levels and explores how lifestyle combinations affect health outcomes.
Project Duration: The study is planned to last for three years. This period will allow us to conduct a thorough analysis of the data and track changes in their health related to lifestyle over time.
Public Health Impact: The findings from this study will be vital for public health initiatives. By identifying which lifestyle changes are most effective at reducing the risk of chronic diseases, health policymakers can design better guidelines and community programs to promote healthier living. This not only improves overall public health but also reduces the economic burden caused by these diseases, such as costs related to medical care and lost productivity. Ultimately, the project will provide evidence-based recommendations that can help people live longer, healthier lives free from chronic diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-lifestyle-and-genetic-risk-with-incidence-of-mental-disorders-and-locomotor-system-diseases

Associations of Lifestyle and Genetic Risk with Incidence of mental disorders and locomotor system diseases

Last updated:
ID:
90923
Start date:
11 November 2022
Project status:
Current
Principal investigator:
Professor Shuiyuan Xiao
Lead institution:
Central South University, China

Stating the aims: This proposed study aims to investigate the hypothesis that adherence to a healthy lifestyle may offset genetic risk for mental disorders or locomotor system diseases and to detect the association between exercise, trauma, and mental status, using the UK Biobank longitudinal data
Scientific rationale: Genetic factors influence the status of mental and locomotor wellness. Increasing evidence indicated that lifestyle habits, especially exercise habits, may relate to genetic factors. Past research has demonstrated that many mental disorders such as depression, anxiety, and schizophrenia have strong, polygenic, genetic bases, as well as many musculoskeletal diseases, such as osteoporosis, osteoarthritis, degenerative spinal diseases, ankylosing spondylitis, scoliosis, et.al. In the last ten years, several genome-wide association studies have identified polymorphisms associated with the development of mental disorders and locomotor system diseases. Lifestyle is a common and crucial modifiable risk factor for them. However, the interactive effects of genetic risk and a healthy lifestyle on this mental and locomotor well-being, as well as the association between these two systems are still not fully appreciated.
Project duration: 36 months
Public health impact: We expect that participants with high genetic risk and unfavorable lifestyle had a synergistic or antagonistic effect on incident mental/ locomotor system diseases compared with participants with low genetic risk and a favorable lifestyle. We expect to obtain the incidence of mental/ locomotor system diseases and their comorbidities, as well as the provision of health care for patients with mental/ locomotor system diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-lifestyle-and-genetic-risk-with-incidence-of-respiratory-diseases

Associations of Lifestyle and Genetic Risk with Incidence of Respiratory Diseases

Last updated:
ID:
84979
Start date:
20 May 2022
Project status:
Current
Principal investigator:
Dr Yan Zhang
Lead institution:
Xiangya Hospital of Central South University, China

Past researchers have demonstrated that genetic factors can increase the risk of respiratory diseases such as asthma, chronic obstructive pulmonary disease (COPD), obstructive sleep apnea hypopnea syndrome (OSAHS), and so on. And several genes have been identified as genetic factors for these respiratory diseases through genome-wide scanning. Moreover, the results of studies based on a large population had shown that the risk of respiratory disease could also be influenced by lifestyle. For instance, smoking is associated with increased risks of COPD, lung cancer, and a high-fat diet is related to the risk of OSAHS and its comorbidities such as cardiovascular diseases and diabetes. However, the potential interaction between genetic variants and lifestyle on these respiratory diseases has not been thoroughly investigated. So, we want to conduct this study to examine that if adherence to a healthy lifestyle can offset genetic risk for respiratory diseases.
We will get the data of genetic information of all participants included in this database and calculate the polygenic risk score across all genetic factors associated with the specific respiratory disease to access the score of genetic risk. And we will also request access to data of lifestyle such as sleep, smoking, and diet of all participants to construct an assessment method of a healthy lifestyle based on risk factors of the specific respiratory disease. After these steps, we will get two different scores of every participant representing genetic risk and lifestyle respectively and could access the interactive effects of genetic risk and healthy lifestyle on specific respiratory diseases based on the two scores.
It will take a long time to build data models for assessing genetic factors and lifestyle, so the estimated duration of this project is 36 months. We expect to find an interaction between genetic risk and lifestyle in respiratory diseases and to remind the public of resisting the genetic risk of respiratory disease by changing specific lifestyles.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-lifestyle-environmental-and-genetic-factors-cerebral-small-vessel-diseases-with-progressive-supranuclear-palsy

Associations of lifestyle, environmental and genetic factors, cerebral small vessel diseases, with progressive supranuclear palsy

Last updated:
ID:
821292
Start date:
15 July 2025
Project status:
Current
Principal investigator:
Miss Yue Yin
Lead institution:
Qilu Hospital of Shandong University, China

1. Investigate gene-environment interactions in PSP susceptibility
PSP is a rare neurodegenerative disease, and clinical evaluation has revealed a predisposition to familial inheritance (Farrell, K., 2024). Environmental factors such as alcohol consumption and exposure to industrial metals also play a role (Litvan, I., 2021). None of these risk factors have been identified as the ultimate cause of PSP. To characterize the combined effects of genetic risk variants and modifiable lifestyle factors (dietary patterns quantified by 24-hour recall, physical activity metrics, and vascular risk profiles) on PSP susceptibility, utilizing polygenic risk scores and gene-environment interaction analyses within UK Biobank’s GWAS data.
2. Elucidate neurovascular mediation mechanisms
High white matter intensity is considered a MRI biomarker for cerebral small vessel disease, while white matter degeneration is characteristic of PSP (Tepedino, M. F., 2024). However, studies on the association between white matter, the presence of vascular risk factors, and disease features in PSP are lacking. To delineate the mediating and moderating roles of CSVD markers, including white matter hyperintensity volume and cerebral microbleed prevalence, in translating genetic/environmental risks into PSP pathogenesis, employing causal mediation analysis with longitudinal adjustment.
3. Identify progression-predictive biomarkers
Diagnosis of PSP remains a major challenge. PSP doesn’t currently include biomarkers in their diagnostic criteria (Giannakis, A., 2025). To determine key prognostic factors influencing PSP progression heterogeneity, focusing on rate of cognitive decline and motor deterioration, through machine learning-driven trajectory clustering and time-to-event modeling.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-lifestyle-environmental-factor-genetic-risk-with-aging-related-diseases

Associations of Lifestyle, Environmental factor, Genetic Risk with Aging-related Diseases

Last updated:
ID:
90060
Start date:
16 September 2022
Project status:
Current
Principal investigator:
Dr Yan Zhang
Lead institution:
Xiangya Hospital of Central South University, China

Past researchers have demonstrated that genetic factors can increase the risk of aging-related diseases such as multiple cancers, cardiovascular and respiratory diseases and so on. Distinct genes have been identified as genetic factors for specific aging-related diseases through genome-wide scanning. Moreover, the results of studies based on a large population had shown that the risk of aging-related diseases could also be influenced by lifestyle and environment. However, the potential interaction between genetic variants, environment and lifestyle on these aging-related diseases has not been thoroughly investigated. So, we want to conduct this study to examine that if adherence to a healthy lifestyle and improvement of environmental situation can offset genetic risk for aging-related diseases.
The estimated duration of this project is 36 months. We expect to find an interaction between genetic risk, environment and lifestyle in aging-related diseases and to remind the public of resisting the genetic risk of aging-related disease by changing specific lifestyles and improving the environment.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-lifestyle-risk-factors-ocular-diseases-and-mental-disorders

Associations of lifestyle risk factors, ocular diseases, and mental disorders.

Last updated:
ID:
93118
Start date:
9 February 2023
Project status:
Current
Principal investigator:
Dr Xiang Jia Zhu
Lead institution:
Fudan University, China

Aims: We aim to determine the specific risk factors for myopia, cataract, and age-related macular degeneration, and further explore the association between these ocular diseases and mental disorders including dementia and depression.
Scientific rationale: Myopia, cataract, and age-related macular degeneration were three main ocular diseases as the leading causes of vision loss worldwide, affecting nearly two-thirds of the populations all over the world. The lifestyles may have some impact on these vision-threatening ocular diseases, and their abnormal visual experience may also result in mental disorders. However, the risk factors for and associations with mental disorders for these vision-threatening ocular diseases remained largely unknown.
Therefore, we will compare baseline characteristics, consumption of caffeine, tea, milk, and oil, outdoor activities, and circadian rhythms in subjects with and without myopia, cataract, and age-related macular degeneration. Furthermore, associations between myopia, cataract, or age-related macular degeneration and incidence of dementia and depression, cognitive test scores, brain magnetic resonanceiImaging (MRI) outcomes, and functional MRI outcomes will be further evaluated.
Project duration: 3 years.
Public health impact: Our work may lead to guidance on lifestyle choices of caffeine, tea, milk, or oil intake, outdoor activity, and day/sleep cycle that will minimize the risk of developing vision-threatening ocular diseases, and also provide information on whether vision-threatening ocular diseases may result in a higher incidence of cognitive impairment and depression.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-lifestyle-sociodemographic-and-nutritional-factors-with-risks-of-non-communicable-diseases-and-mortality

Associations of Lifestyle, Sociodemographic, and Nutritional Factors with Risks of Non-Communicable Diseases and Mortality

Last updated:
ID:
235206
Start date:
27 November 2024
Project status:
Current
Principal investigator:
Dr Linglong Peng
Lead institution:
Chongqing Medical University, China

Non-communicable diseases and mortality pose major global disease burdens. Increasing studies have explored associations between non-communicable diseases, mortality, and influencing factors using epidemiological methods like cohort studies and causal inference approaches such as Mendelian randomization. Early interventions for non-communicable diseases can effectively improve prognosis, reduce disease burden, and lower mortality risk. Hence, this project aims to elucidate causal relationships between risks of non-communicable diseases (including hypertension, stroke, coronary heart disease, diabetes, dementia, Alzheimer’s disease, Parkinson’s disease, schizophrenia, depressive disorders, chronic obstructive pulmonary disease, cancer) and mortality with candidate risk factors encompassing lifestyle (e.g., physical activity, sedentary behavior, sleep, smoking, alcohol), sociodemographic characteristics (e.g., gender, age, education, poverty), and nutrition (e.g., nutrients, dietary patterns). Objectives are: 1) explore associations of lifestyle factors with non-communicable disease and mortality risks, 2) investigate sociodemographic impacts on these risks, 3) evaluate nutrition factor relationships with these risks, 4) provide novel index, statistical methods, and predictive models from lifestyle, sociodemographic, and nutritional perspectives to reduce global non-communicable disease and mortality burdens. The research will utilize appropriate statistical methods to uncover the associations or pathways between the aforementioned factors and non-communicable diseases, spanning a duration of 36 months. We aim to significantly reduce the global burden of non-infectious diseases and lower mortality risks. This comprehensive approach underscores our commitment to advancing the understanding of chronic diseases through a multifaceted analysis of lifestyle, sociological, and nutritional impacts, ultimately contributing to the development of effective intervention strategies and public health policies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-multi-omics-lifestyles-environmental-and-other-factors-in-the-occurrence-progression-and-prognosis-of-metabolic-cardiovascular-and-cerebrovascular-diseases

Associations of multi-omics, lifestyles, environmental, and other factors in the occurrence, progression, and prognosis of metabolic cardiovascular and cerebrovascular diseases.

Last updated:
ID:
188040
Start date:
5 November 2024
Project status:
Current
Principal investigator:
Dr Zi Ye
Lead institution:
Wannan Medical College., China

Metabolic cardiovascular diseases are the most common cause of morbidity and mortality worldwide, representing a major public health challenge. Some clinical, genetic, and environmental risk factors related to metabolic cardiovascular diseases have been well-established, such as obesity, diabetes, cigarette smoking, air pollution exposures, diet and physical activity. However, research on metabolic cerebrovascular diseases is still insufficient. Recent studies suggested that muti-omics (including metabolomics, proteomics, and radiomics) imbalance also played an important role in the development and progression of diseases. Combining multi-omics data with clinical, genetic, lifestyle, and environmental risk factors might help to improve the evaluation of the risk of metabolic cardiovascular and cerebrovascular diseases. Moreover, exploring the independent and joint effect of multi-omics, genetic, lifestyle, and environmental risk factors on the morbidity and mortality of metabolic cardiovascular and cerebrovascular diseases will help to assess the modifiable risk, take strategies to prevent the metabolic cardiovascular and cerebrovascular diseases, and reduce the mortality. In this project, we aim to investigate the associations of multi-omics, genetic, lifestyle and environmental risk factors as well as their interactions with the morbidity and mortality of metabolic cardiovascular and cerebrovascular diseases; to evaluate the modification effect of genetic susceptibility on above associations; and to explore biomarkers that may play a role in the occurrence, progression, and prognosis of metabolic cardiovascular and cerebrovascular diseases. Given that cognitive impairment is closely related to the occurrence of cerebrovascular diseases, we will also focus on the role of above associations in the occurrence of cognitive impairment.
Our project is expected to last 36 months, and we will endeavor to contribute to the knowledge surrounding the impact of multi-omics, genetic, lifestyle, and environmental risk factors on the morbidity and mortality of metabolic cardiovascular and cerebrovascular diseases and cognitive impairment, and therefore improve health status and extend healthspan.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-oral-and-gut-microbiome-related-exposures-with-cancer-risk-and-mortality

Associations of oral and gut microbiome-related exposures with cancer risk and mortality

Last updated:
ID:
52576
Start date:
18 September 2019
Project status:
Closed
Principal investigator:
Dr Emily Vogtmann
Lead institution:
National Cancer Institute, United States of America

Both the oral and gut microbiome (i.e., the collection of microbes including bacteria, archaea, fungi, viruses, and other eukaryotic microbes found in the oral cavity or the gut) are likely to be involved in the development of leading causes of mortality in developed countries, such as cardiovascular disease, diabetes, and cancers; however, associations between the microbiome and cancer or mortality risk remain unclear. This is mostly due to the lack of large prospective studies with oral and/or fecal samples available for the assessment of the microbiome with adequate follow-up to observe microbiome-disease associations. In the meantime, it is informative to assess associations of established microbiome-related exposures with cancer and mortality risk to provide a better understanding of the mechanisms underlying microbiome-disease associations. Therefore, our objective is to investigate the associations of exposures related to oral and gut microbiome composition with overall and site-specific cancer risk, as well as overall and cause-specific mortality in the large, prospective UK Biobank study. We will characterize oral microbiota-related metrics by assessing oral health, including history of mouth ulcers, gingivitis and periodontal disease, loose teeth, dental caries, and denture use. Exposures related to the gut microbiome will include long-term antibiotic use, birth by cesarean section, inflammatory and irritable bowel syndromes, Celiac disease, appendicitis, and average number of bowel movements. To assess these associations, we will use statistical models which take into account variables that may affect the microbiome and/or cancer risk and mortality, such as smoking, alcohol consumption, and diet. These findings could provide evidence and further insight into the role of exposures related to the microbiome in cancer risk and premature mortality, and provide evidence for the continuing exploration into the etiology of microbial-related health states. Findings from this study may also provide insight into prevention strategies and translate into public health and clinical recommendations for oral and gut health.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-physical-activity-and-physical-function-with-the-risk-of-covid-19-infection-hospitalisation-and-death

Associations of physical activity and physical function with the risk of COVID-19 infection, hospitalisation, and death.

Last updated:
ID:
70184
Start date:
9 February 2021
Project status:
Closed
Principal investigator:
Mr Malik Hamrouni
Lead institution:
Loughborough University, Great Britain

Physical inactivity is associated with lowered immune function, higher levels of inflammation and poorer cardiovascular and metabolic health. Collectively, this may increase an individual’s susceptibility to COVID-19 and their risk of developing severe and/or fatal health complications during the course of infection. Despite this, there is limited research exploring a protective role of physical activity against COVID-19 infection, particularly in at risk populations such as older adults and individuals with obesity.

Old age and obesity are linked with greater risk of COVID-19 hospitalisation and death. This may be partly due to the negative effects of aging and fat accumulation on inflammation and cardiovascular and metabolic health. However, increasing levels of physical activity may lessen these effects, as older adults and individuals with obesity who are physically active have been shown to have lower levels of inflammation and improved cardiovascular and metabolic health compared to their less active counterparts. Importantly, this suggests the potential for regular physical activity to reduce the burden of COVID-19 typically observed in such populations.

Therefore, this study aims to investigate the relationship between physical activity level and COVID-19 infection risk, hospitalisation and death, firstly independent of age and fatness, and then across different age groups and obesity categories. Moreover, as the health benefits of physical activity may occur largely through improving fitness and muscular strength, we also aim to provide insight into whether measures of physical function, such as walking speed and hand grip strength, may also be linked with COVID-19 outcomes. Further analysis will be undertaken to determine whether any protective associations of physical activity and/or physical function may be explained by certain health markers.

The findings from this project will provide evidence to support increased public health efforts to promote physical activity as a feasible and achievable lifestyle habit to reduce the national burden of COVID-19. The proposed research will also inform future management and prevention strategies for other common respiratory infections and/or subsequent coronavirus outbreaks. The project is estimated to last 12 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-physical-activity-with-chronic-diseases-cardiorespiratory-function-and-cognitive-function

Associations of physical activity with chronic diseases, cardiorespiratory function, and cognitive function

Last updated:
ID:
781059
Start date:
17 June 2025
Project status:
Current
Principal investigator:
Mr Junyu Wu
Lead institution:
Shanghai University of Sport, China

Research Title:
Associations of physical activity with chronic diseases, cardiorespiratory function, and cognitive function

Research Questions:

What is the relationship between physical activity and the incidence and progression of chronic diseases (e.g., cardiovascular disease, diabetes, osteoarthritis)?

How does physical activity influence cardiorespiratory function and cognitive performance in adults, particularly in aging populations?

Do cardiorespiratory and cognitive functions mediate the relationship between physical activity and chronic disease outcomes?

Objectives:

To examine the association between physical activity levels and the risk of developing or dying from chronic diseases.

To investigate the impact of physical activity on cardiorespiratory fitness and cognitive function.

To assess whether cardiorespiratory and cognitive functions act as mediators in the pathway linking physical activity to chronic disease prevention.

To provide evidence supporting targeted health promotion strategies involving physical activity.

Scientific Rationale:
Chronic diseases are leading contributors to global disability and mortality, especially in aging populations. Physical activity has been shown to influence multiple physiological systems, including cardiovascular and respiratory health, as well as cognitive functioning. However, the complex relationships between these domains remain insufficiently understood. By leveraging large-scale population data, this research will clarify the mechanisms by which physical activity confers protective effects against chronic conditions, ultimately informing preventative strategies and promoting healthy aging.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-physical-function-with-incidence-of-chronic-conditions-and-risk-of-all-cause-and-specific-mortality-longitudinal-analyses-accounting-for-genetic-predisposition

Associations of physical function with incidence of chronic conditions and risk of all-cause and specific mortality: longitudinal analyses accounting for genetic predisposition

Last updated:
ID:
98633
Start date:
26 April 2023
Project status:
Current
Principal investigator:
Dr Rubén López-Bueno
Lead institution:
University of Valencia, Spain

We will examine to what extent physical activity and muscular strength contribute to reduce the incidence of cardiovascular, cancer and other common chronic conditions when accounting for genetic predisposition. To date, most of scientific research has observed strong inverse associations, but recent research has suggested that these might be of lesser extent that commonly expected. Our research will provide more accurate and updated insights on this topic.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-physical-multimorbidity-patterns-and-heart-structure-with-cognition-and-neuroimaging-outcomes

Associations of physical multimorbidity patterns and heart structure with cognition and neuroimaging outcomes

Last updated:
ID:
97089
Start date:
20 September 2023
Project status:
Current
Principal investigator:
Professor Shaojun Tang
Lead institution:
The Hong Kong University of Science and Technology (Guangzhou), China

The project will study the potential associations of physical multimorbidity (such as chronic low back pain, Parkinson’s disease, stroke and knee osteoarthritis), heart structure, lung function and active exercise with cognitive functions. To explain further, physical activity means the time or intensity of daily activity, or other physical performances. Lung functions and heart structure indicate the healthy condition of cardiopulmonary function while cognitive functions mean the variety of mental processes, including memory and decision making. Based on previous studies, the morbidity of cardiometabolic multimorbidity and the risk of incident arrhythmias (which is related to dementia) is related to physical activity. Lung function is known to be influenced by 391 genes, and related to a variety of chronic diseases. The heart structure anomaly can be caused by genes including the GATA family, the MEF2 family and the NK family. Therefore, it is rational to hypothesise that multimorbidity, intensity of daily activity, heart structure and lung health are related to the happening or development of disorder of cognitive functions and the effects are modified by genotypes.
The duration of this project is approximately 3 years. The results of this project will indicate the potential associations between specific genotypes, differentially expressed genes, physical multimorbidity, heart structure, lung function and active exercise and cognitive functions. Therefore, these findings could tell people about possible causes or factors of the development of dementia and extensive brain imaging differences, thereby providing more information for clinicians and therapists to prevent the happening and decrease the risk of these disorders.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-serum-biomarkers-with-chronic-obstructive-pulmonary-disease-risk-and-survival

Associations of serum biomarkers with chronic obstructive pulmonary disease risk and survival

Last updated:
ID:
84525
Start date:
18 March 2022
Project status:
Current
Principal investigator:
Dr Xikang Fan
Lead institution:
Jiangsu Provincial Center for Disease Control and Prevention, China

Chronic Obstructive Pulmonary Disease (COPD) is the leading cause of morbidity and mortality worldwide. Major risk factors for COPD include tobacco smoking, age, sex, lung growth and development, occupational and environmental exposure, socioeconomic status. Despite the well-established epidemiologic evidence, however, the exact mechanisms involved remain inconclusive. Meanwhile, increasing data suggest a critical role of serum metabolic alterations in relation to COPD. Although these previous studies have indicated the associations between serum biomarkers (e.g., IGF-1, vitamin D, sex hormones, lipid profiles, etc.) with COPD, the data remain limited and large-scale prospective population studies are lacking. Therefore, to better understand the role of metabolic alterations in the development and progression of COPD, a detailed investigation of serum biomarkers with COPD is needed. Moreover, prevention and/or early detection can effectively reduce the burden caused by COPD. Risk prediction models can be used to assess individual risk. So far, most risk prediction models for COPD often include clinical indices with only very limited basic epidemiological factors such as age, sex, tobacco smoking, etc. In this study, we will utilize the data from UK biobank to prospectively investigate associations of serum biomarkers with COPD risk and survival. We will use the data to develop a model to predict each participant’s risk of developing COPD, according to their different combinations of risk factors. This research will make important contributions to the guidance of personalized prevention of COPD.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-sleep-duration-with-the-incidence-of-severe-non-alcoholic-fatty-liver-diseasenafld

Associations of sleep duration with the incidence of severe non alcoholic fatty liver disease(NAFLD).

Last updated:
ID:
409444
Start date:
23 October 2024
Project status:
Current
Principal investigator:
Miss Qian Wang
Lead institution:
Southern Medical University, China

Non alcoholic fatty liver disease (NAFLD) which affects 25% of the population worldwide is rapidly becoming the most common cause of chronic liver disease and its prevalence is expected to increase in the near future.Obviously, NAFLD is one of the diseases that seriously affects human health, with a high incidence rate, many and complex pathogenic factors, and there is currently no approved drug treatment, and the first-line treatment for NAFLD is lifestyle modification. Therefore, it is particularly important to identify all modifiable lifestyle factors associated with the onset of NAFLD.As sleep occupies 1/3 of the human life cycle, it is of great value and significance to study its association with severe non-alcoholic fatty liver disease. At the same time, sleep problems have become the most common medical and health problems in today’s society, and it is urgent to study its association with severe non-alcoholic fatty liver disease.
The association between sleep duration and NAFLD was unclear.We aim to explore the associations between sleep duration and NAFLD in a large population-based cohort study. Our project will provide evidence for developing efficient strategies to reduce the morbidity of NAFLD. This project is expected to last for 36 months. The findings may deepen the understanding of associations between sleep duration and NAFLD, and may provide strong evidence for the prevention of NAFLD, and even make a significant contribution to global public health.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/associations-of-workplace-enviroment-with-cardio-cerebral-vascular-disease-diseases-and-their-interactions-with-genetic-susceptibility-and-lifestyle

Associations of workplace enviroment with cardio-cerebral vascular disease diseases and their interactions with genetic susceptibility and lifestyle

Last updated:
ID:
161956
Start date:
9 January 2025
Project status:
Current
Principal investigator:
Mr Hongyu Liu
Lead institution:
The Second Affiliated Hospital of Nanchang University, China

This project aimed to investigate the relationship between exposure to workplace environment pollution (e.g., chemical smoke, smoking, noise, automobile exhaust) and selected chronic diseases, as well as the potential interactions between these workplace environment factors and individual genotypes and lifestyle factors (e.g., dietary habits, physical activity, sleep patterns). Another goal is to clarify the relationship between workplace environmental exposure and multiple diseases. We will focus on cardio-cerebrovascular and respiratory diseases (e.g., atrial fibrillation, heart failure, hypertension, coronary heart disease, stroke, Alzheimer’s disease, chronic obstructive pulmonary disease) and their related characteristics (e.g., cardiac function, pulmonary function, etc.).
Specifically, we aimed to 1) assess the effects of various workplace environmental exposures, including chemical fumes, smoke, noise, and diesel exhaust, on i) cardiovascular outcomes; ii) neurological outcomes; iii) respiratory prognosis; iv) other health outcomes of interest to us; 2) explore potential interactions among these different workplace environmental exposures; 3) identify individuals with a genetic predisposition and the ways in which the health effects of workplace environment exposures can be modulated by individual genetic variation; 4) examine whether lifestyle factors (e.g., dietary habits, physical activity, sleep patterns) play a potential role.
The program will be divided into subprograms focused on a few selected health outcomes (e.g., atrial fibrillation, heart failure, coronary heart disease, stroke, Alzheimer’s disease, chronic obstructive pulmonary disease) and is expected to last approximately 3 years. This study will contribute to a better understanding of the interaction between different modifiable workplace environment factors and genetic susceptibility, hopefully facilitating more effective prevention strategies and providing new evidence for scientific and rational workspace layout. This project may lead to more comprehensive personalized approaches to prevention and treatment, taking into account both individual-level factors and social-level factors in the workplace environment.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assortative-mating-in-the-uk-biobank-intergenerational-health-implications

Assortative mating in the UK Biobank: intergenerational health implications

Last updated:
ID:
52286
Start date:
17 August 2020
Project status:
Current
Principal investigator:
Mr Thomas Michael Mello Versluys
Lead institution:
Imperial College London, Great Britain

This project explores genetic and phenotypic (i.e., non-genetic traits) similarities in human couples for a diverse range of genetic and phenotypic traits. Mating with similar individuals is described as assortative mating (AM), and there is considerable debate about the health consequences of this behaviour. Current empirical tests of this are contentious, so further research is required. This project tests whether AM is associated with fertility (i.e., number of children born) and offspring (viability (i.e., child health) in a novel population. It does this over multiple generations, testing not just whether offspring are affected by parental AM, but whether those offspring themselves go on to be affected. In doing so, it provides insights into whether AM could lead to the intergenerational transmission of health risk.

The scientific and health implications of this are considerable. The causes and consequences of AM have been the subject of intense academic interest and speculation for decades (Robinson et al. 2017; Abdellaoui et al. 2014; Bereczkei & Csanaky 1996; Thiessen & Greg 1980). If the behaviour is associated with high fitness (e.g., improved fertility), it might suggest an adaptive function (i.e., the it was selected for during human evolution). If the opposite is true, it will raise questions about how the behaviour has been maintained, potentially implicating sociocultural factors. Understanding the relationship between AM and offspring health could be important for medical researchers, but it is also of public-health interest. For example, it has been suggested that AM for body-mass-index has led to the intergenerational transmission of overweight, contributing to the “obesity crisis” in the developed world (Speakman et al., 2007). In terms of duration, the project is expected to last until 2022-23.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/assortative-mating-in-the-uk-population

Assortative mating in the UK population

Last updated:
ID:
19127
Start date:
8 May 2016
Project status:
Current
Principal investigator:
Dr David Hugh-Jones
Lead institution:
University of East Anglia, Great Britain

This project looks at whether people are genetically similar to their partners. This will help us
understand two things.

First, how inequality is transmitted across generations. If advantaged people form households with others like them, they can transmit extra advantage to their children. Some of this advantage may be genetic – for example, height is partly genetic, and tall people do well in the labour market (Case et al. 2006).

Second, how ‘well-mixed’ the population is genetically. This is important for researchers estimating the heritability of health conditions: statistical models of heritability depend on a well-mixed population. Our research will:

(i) Improve illness diagnosis and treatment by allowing more accurate estimates of heritability which take account of population structure. Non-random mate choice influences the genetic make-up of a population, and understanding these fundamental processes would aid in improving quantitative genetics and molecular genetics research.

(ii) Improve the promotion of health throughout society by helping us to understand the transmission of social inequality. Inequality has risen in the UK and elsewhere in recent decades, and may be a major cause of poor health outcomes (Marmot et al. 1991; Wilkinson et al. 2011). We will look at how cohabiting couples correlate on variables including education level, height and income. We will test for differences in correlation across age groups.

Then, using genomic data, we will measure couples? genetic similarity. Again, we will compare these measures of similarity across age groups.

We will also look specifically at whether couples have similar genes linked to height and education – two traits where `like marries like`. All participants for whom genetic data exists for both partners cohabiting in a household.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/asthma-a-test-case-for-precision

Asthma-a test case for precision

Last updated:
ID:
61031
Start date:
9 June 2020
Project status:
Closed
Principal investigator:
Justin O'Sullivan
Lead institution:
University of Auckland, New Zealand

Asthma is a very common disease in children and adults. Lots of small changes in our DNA have been linked to our chances of developing Asthma. However, what these small DNA changes do and how they contribute to your chances of developing asthma is still unknown. Asthma looks different in different people. This makes it very difficult to say what form of asthma a person has. Therefore, it is hard to group patients and understand how asthma works. As a result, current treatment for asthma is essentially optimized using a trial-by-error search for a suitable treatment for a patient. Because of this, asthma has enormous public healthcare and social costs.

In this study, we will identify the genes that are affected by the small DNA changes that have been linked to our chances of developing asthma. We will combine information on how DNA folds in cells with information on gene expression for each DNA sequence in the UkBioBank. Linking all this information with the genotypes and asthma status of people in the UK Biobank, will enable us to identify asthma subtypes and the key changes that put individuals at risk of developing asthma. This will mean we can identify biological pathways for the development of drug targets for asthma. We will also develop new and improved ways to group people with asthma. This will change the ways we apply precision therapy for asthma patients.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/at1r-as-the-core-bond-between-aml-and-cvd

AT1R as the core bond between AML and CVD

Last updated:
ID:
106460
Start date:
5 April 2024
Project status:
Current
Principal investigator:
Dr Yi Pan
Lead institution:
University of Missouri, United States of America

Acute Myeloid Leukemia (AML) and Cardiovascular Disease (CVD) have significant clinical association. To determine if there is a shared target between AML and CVD, we will analyze the genetic pattern between AML and CVD. We expect angiotensin 2 receptor type 1 (AT1R) to be the core bond between AML and CVD. AT1R has been implicated in various CVDs, such as hypertension, coronary artery disease and stroke. However, AT1R’s role in AML has not been well studied. In our preliminary studies, we found AT1R expression was highly increased on the cell surface of human AML cells compared with healthy control, and genetic mouse models showed a delay of leukemia development after AT1R knockout.
Our aim is to identify shared targets between AML and CVD and evaluate AT1R’s role in AML within 36 months.
These studies should provide a strong rationale in support of AT1R inhibition as a potential treatment for human AML. Moreover, standard chemotherapy regimens used for AML treatment are associated with significant side effects including cardiotoxicity. Studies have shown that angiotensin signaling is important for chemotherapy-induced cardiotoxicity. AT1R inhibition may also reduce cardiotoxicities in AML patients undergoing chemotherapy.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/attributable-risk-of-genetic-lifestyle-and-environmental-factors-on-the-incidence-and-mortality-of-pan-cancer

Attributable risk of genetic, lifestyle and environmental factors on the incidence and mortality of pan-cancer

Last updated:
ID:
82535
Start date:
25 March 2022
Project status:
Current
Principal investigator:
Professor Hongbing Shen
Lead institution:
Nanjing Medical University, China

Aims: To investigate the associations between genetic, lifestyle, environmental factors and incidence and mortality of pan-cancer.

Scientific rationale: site-specific cancers are known to share common risk factors, including genetic, lifestyle and environmental factors. Many lifestyle and environmental risk factors have been identified, including cigarette smoking, air pollution, occupational exposures, diet and physical activity. Genetic risk was evaluated partially using polygenic risk score for chronic diseases and evidence showed that combining polygenic risk score with rare mutaions help to improve the evaluation of the genetic risk. Understanding the independent and joint effect of genetic, lifestyle and environmental factors on the incidence and mortality of cancer will help to assess the modifiable risk and take strategies to prevent the cancer and reduce the mortality

Project duration: 36 months

Public health impact: This project is expected to improve understanding of the impact of genetic, lifestyle, environmental factors on the incidence and mortality of cancer and therefore contribute to improving health status and extending healthy lifespan.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/attributable-risk-of-multi-omics-genetic-lifestyle-and-environmental-factors-on-the-morbidity-and-mortality-of-cardio-metabolic-diseases

Attributable risk of multi-omics, genetic, lifestyle and environmental factors on the morbidity and mortality of Cardio-metabolic diseases

Last updated:
ID:
95387
Start date:
23 January 2023
Project status:
Current
Principal investigator:
Dr Yan Liu
Lead institution:
Sun Yat-Sen University, China

Aims: To investigate the associations of multi-omics, genetic, lifestyle and environmental risk factors with the morbidity and mortality of cardio-metabolic diseases.
Scientific rationale: Cardio-metabolic diseases are the most common cause of morbidity and mortality worldwide, representing a major public health challenge. Some clinical, genetic, and environmental risk factors associated to cardio-metabolic diseases have been well-established, including hypertension, diabetes, obesity, cigarette smoking, air pollution exposures, diet and physical activity. Recent studies suggested that muti-omics (including metabolomics, proteomics, and radiomics) imbalance also played an important role in the development and progression of cardio-metabolic diseases. Recent evidence showed that combining multi-omics data with clinical, genetic, lifestyle, and environmental risk factors might help to improve the evaluation of the risk of cardio-metabolic diseases. Understanding the independent and joint effect of multi-omics, genetic, lifestyle, and environmental risk factors on the morbidity and mortality of cardio-metabolic diseases will help to assess the modifiable risk and take strategies to prevent the cardio-metabolic diseases and reduce the mortality. Machine learning techniques are being increasingly adapted for use in the risk-prediction failed. This study also investigated the predictive values of above risk factors for cardio-metabolic diseases-related long-term outcomes by the machine learning techniques.
Project duration: 36 months
Public health impact: This project is expected to improve understanding of the impact of multi-omics, genetic, lifestyle, and environmental risk factors on the morbidity and mortality of cardio-metabolic diseases, and therefore contribute to improving health status and extending healthy lifespan.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/auditory-multiomics-in-the-uk-biobank

Auditory Multiomics in the UK Biobank

Last updated:
ID:
74463
Start date:
16 April 2025
Project status:
Current
Principal investigator:
Dr Christopher R. Cederroth
Lead institution:
University of Tübingen, Germany

Tinnitus is a constant buzz in the ear or in the head, often resulting from a compensation to reduced auditory input (i.e. hearing loss). While it is common in the population, nearly 2% suffer from it to a debilitating degree and what makes it severe is currently not well understood. However, this knowledge is important for developing treatments targeting tinnitus in the group of people who seek help.

We have recently shown that severe tinnitus is more likely prone to genetics than any other subtype, which emphasizes the need to conduct genetic, epidemiology, neuroimaging and biomarker studies in this group. We have also conducted a large case/control protein screen on plasma samples and identified 5 biomarkers associated with constant tinnitus. Our aim is to combine the four into a multi-omic approach that will lead to a better understanding of how genes i) relate to brain alterations leading to severe tinnitus, ii) interact with environmental factors to increase severity, and iii) impact the blood proteome to increase severity.

In order to better map the mechanisms associated with severe tinnitus, we will also investigate other disorders highly associated with tinnitus (e.g. hearing loss, depression, anxiety, pain) and dissociate tinnitus from these comorbidities. Parkinson’s, which is unrelated to tinnitus, will serve as a negative reference group. We will use established methods to search for genetic variations associated with severe tinnitus, and then test whether these genetic factors are associated with specific changes in brain structure and function, environmental factors, and blood biomarkers. The project duration is 3 years, and is based on already acquired data.

Thus far, a multi-omic approach to severe tinnitus is lacking and will provide a significant advance in knowledge for a rare condition for which optimized treatment options are needed. Only a large biobank such as the UK Biobank may help provide insights into the mechanisms of severe tinnitus. The findings from this study will provide a basis for pre-clinical research to increase fundamental knowledge of this disorder, and it will improve patient stratification (subtyping) based on genetic and neural markers which could be used for personalized medicine, i.e. to direct an individual patient toward the most appropriate treatment for them.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/augmenting-data-driven-discovery-of-disease-risk-genes-using-multivariate-and-integrative-approaches-to-genome-wide-association-analysis

Augmenting data-driven discovery of disease risk genes using multivariate and integrative approaches to genome-wide association analysis

Last updated:
ID:
27386
Start date:
17 July 2018
Project status:
Current
Principal investigator:
Professor Xin He
Lead institution:
University of Chicago, United States of America

The ultimate aim of this project is to draw new connections between genes and diseases, such as psychiatric disorders and diabetes. Our group develops methods and software for discovering gene-disease associations from large-scale data. We take two distinctive approaches to this problem: we develop multivariate methods that generate informative links between genes and multiple phenotypes?disease, biomarkers and environmental exposures; we integrate external information on genetic variants, such as gene expression (eQTLs), to guide discovery of disease risk genes. The UK Biobank offers a unique opportunity to apply these approaches and expand biological insights into disease risk. The UK Biobank has collected health, medical and genetic data at an unprecedented scale and scope. It would be of great value to the UK Biobank initiative, we believe, to help researchers better understand the genetic basis of complex inherited diseases. The proposed research will employ highly innovative methods to analyze the genetic data. By developing, applying and evaluating these methods on the UK Biobank data, then disseminating them to the larger research community as standalone software packages, we believe these efforts will help to realize the true potential of UK Biobank Project. Our research efforts are roughly broken down into 5 stages: (1) we download the UK Biobank data, and take `quality-control` steps to manipulate data that may compromise robustness of our results; (2) we develop preliminary software implementations using programming languages such as R; (3) we test our software in smaller data sets to ensure accuracy and reproducibility; (4) we apply our methods to the Biobank data, then interpret and verify the results, often with the aid of other bioinformatics resources; and (5) we develop and disseminate user-friendly software toolkits. We have accumulated experience in all these research stages. Full cohort.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/autoimmune-diseases-and-cardiovascular-risks-investigating-possible-pathophysiological-mechanisms-and-disease-specific-risk-prediction-tools

Autoimmune diseases and cardiovascular risks. Investigating possible pathophysiological mechanisms and disease-specific risk prediction tools.

Last updated:
ID:
703810
Start date:
7 May 2025
Project status:
Current
Principal investigator:
Professor Werner Budts
Lead institution:
Katholieke Universiteit Leuven, Belgium

Background: Individuals with autoimmune diseases carry elevated risk for cardiovascular disease similar to the risk brought by type 2 diabetes. Yet the exact underlying biological mechanisms remain unclear, and currently available models are insufficient to accurately predict cardiovascular risks in these patients.
Objectives: To i) determine the extent of the association between autoimmune diseases and cardiovascular risk that is explained by known cardiovascular risk factors; ii) examine possible underlying biological mechanisms beyond known cardiovascular risk factors by investigating how blood biomarkers mediate observed associations; iii) develop a risk prediction model specifically for individuals affected by autoimmune disorders, considering both traditional and disease specific risk factors.
Methods: The study will use one of the largest and richest datasets available with cardiovascular risk factors, blood biomarkers and imaging data, the UK Biobank. Analyses will consider 19 of the most common autoimmune diseases and 12 cardiovascular outcomes.
Expected outcomes: A better understanding of possible pathophysiological mechanisms underlying cardiovascular risks in patients with autoimmune diseases may support the future development of preventive treatments. Tools for accurate identification of individuals at highest risk of developing cardiovascular events early in life may further support more targeted and timely risk modification interventions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/automated-actigraphy-analysis-for-identification-of-prodromal-parkinsons-disease-or-related-disorders

Automated actigraphy analysis for identification of prodromal Parkinson’s disease or related disorders

Last updated:
ID:
97043
Start date:
22 March 2023
Project status:
Current
Principal investigator:
Dr Emmanuel During
Lead institution:
Icahn School of Medicine at Mount Sinai, United States of America

In many cases, Parkinson’s disease (PD) and related disorders are preceded by early sleep disturbances. A characteristic sleep disorder is REM sleep behavior disorder (RBD), which is an abnormal motor disinhibition during REM sleep, causing twitches and dream-enactment episodes. In at least 80 % of RBD cases, PD or a related disorder will manifest within 10 years. The prevalence of RBD is between 1-2% in the general population. An effective screening method of RBD would have high impact in terms of identifying patients at PD risk, when progression of their neurodegenerative disease can be slowed down or halted.
Current screening methods use questionnaires; however, despite their good accuracy, close to 90%, RBD questionnaires do not have sufficient precision for general population screening. Moreover, the gold standard for a definite diagnosis is costly and requires an in-lab sleep test. Our research suggests that actigraphy could be used on its own or as a supplement to questionnaires for screening purposes. Using machine learning, we found that actigraphy could classify RBD correctly in 92.9 % of recordings. Moreover, this model supplies RBD classification scores that indicate disease severity.
In this project, associations between actigraphy-based RBD classification scores, genetics, and various clinical variables of interest. Carrying out this study in the UK Biobank has many advantages to previous studies of RBD, as available datasets based on definite diagnosis are very scarce. Applying our detection algorithm in the UKBB may allow to find new genotypes and phenotypes (described in detail in A4) related to RBD and disease progression.
During this 3-year project, we will: 1) Fine-tune the machine learning-based classifier for data in the UK; 2) Run the RBD classifier on all accelerometer data from suitable participants; 3) Replicate genetic profiles known for PD and definite RBD, in individuals predicted to have RBD using our method; 4) Describe cognitive, motor, autonomic, and biological differences in individuals predicted to have RBD using our method, against individuals predicted to not have RBD
The public health impact of this research would be: 1) to increase our understanding of PD subtypes and disease mechanisms, 2) accelerate discovery of therapies for PD by enhancing recruitment pipelines with the inclusion of participants with prodromal PD, ie with RBD, and 3) offer a screening tool for RBD once neuroprotective therapies for PD become available.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/automated-analysis-of-abdominal-aortic-calcification-in-dxa-vfa-images

Automated analysis of abdominal aortic calcification in DXA VFA images

Last updated:
ID:
35900
Start date:
2 November 2018
Project status:
Closed
Principal investigator:
Mr Luke Anthony Chaplin
Lead institution:
University of Manchester, Great Britain

Cardiovascular diseases, such as strokes and heart attacks, are among the most common causes of death in the UK. These diseases are caused by a build of fat in the walls of the blood vessels near the heart. When this fat continues to develop, calcium starts to build up as well. Calcium in the walls of the aorta, abdominal aortic calcification (AAC), can be seen on x-ray images and is an indicator of similar build-ups in smaller vessels around the heart. Dual-energy x-ray absorptiometry (DXA) images are x-ray images taken to check for bone thinning in older people, they incidentally show this AAC and can be used to measure it. This is time consuming for radiologists and so not consistently done.

This project aims to teach a computer to measure the AAC and use it to better predict who is at risk of having a heart attack or stroke so that treatment can be started earlier for these people. The computer will be tested by getting it to predict the scores that a radiologist would give for the same images. Once the computer can match the scores of a human, then it can be trained to do better than a human by testing it to predict which people have had heart attacks or strokes since the scan.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/automated-and-robust-cardiac-function-quantification-from-cardiac-mri

Automated and robust cardiac function quantification from cardiac MRI

Last updated:
ID:
93224
Start date:
11 October 2022
Project status:
Closed
Principal investigator:
Mr Cosmin-Andrei Hatfaludi
Lead institution:
Transilvania University, Romania

Cardiovascular disease is the leading cause of death globally, according to the World Health Organization. Cardiovascular magnetic resonance imaging (MRI) is considered the gold standard for evaluating heart function. Estimating the ventricular end-systolic (ESV) and end-diastolic (EDV) volumes, stroke volume (SV) and ejection fraction (EF) from cardiac MRI is a prerequisite for assessing cardiovascular diseases, and typically requires careful and precise contouring of the ventricles. Although having been the subject of intense research over the years, cardiac function quantification from MRI is still not a fully automatic process in clinical practice. This is partly due to the shortage of training data covering all relevant cardiovascular disease phenotypes. We propose to develop a deep learning convolutional neural network to predict the end-diastolic volume, end-systolic volume, and implicitly the ejection fraction from cardiac MRI without explicit segmentation. The regression network will take as input short axis images and directly predict ESV, EDV, SV and EF. Depending on the results we will likely resort to the generation of synthetic to expand the available data sets that consist of predominantly healthy subjects to include more cases with reduced ejection fraction.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/automated-cardiac-mri-reporting-and-disease-classification-using-large-language-models-llms

Automated Cardiac MRI Reporting and Disease Classification using Large Language Models (LLMs)

Last updated:
ID:
475772
Start date:
5 August 2025
Project status:
Current
Principal investigator:
Professor Lequan Yu
Lead institution:
University of Hong Kong, Hong Kong

Research Questions:
1. How can large language models (LLMs) be utilized to automate the generation of cardiac MRI reports while ensuring accuracy and consistency with clinical experts?
2. What robust machine learning models can be developed to improve the accuracy and timeliness of cardiac disease classification from CMR data?
3. How can these advancements reduce the diagnostic workload on clinicians while enhancing patient outcomes?

Research Objectives:
1. To develop an automated system using machine learning and LLMs for generating detailed cardiac MRI reports, thereby increasing diagnostic efficiency.
2. To create and validate machine learning models that accurately classify cardiac diseases from CMR data, facilitating earlier and more precise interventions.
3. To evaluate the impact of these technologies on reducing clinician workload and improving diagnostic accuracy and patient prognosis.

Scientific Rationale:
Cardiac magnetic resonance imaging (CMR) is the gold standard for assessing cardiac function and diagnosing cardiovascular diseases. However, the complexity of CMR interpretation requires significant expertise, leading to potential variability in diagnoses and substantial time investment from clinicians. By integrating advanced machine learning and LLM techniques, this research aims to streamline the diagnostic process, ensuring more consistent and accurate reporting. This approach not only alleviates the burden on healthcare professionals but also enhances the overall quality of patient care through timely and precise diagnoses. The proposed research has the potential to significantly advance the field of cardiac imaging, contributing to better health outcomes and more efficient clinical workflows.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/automated-cardiovascular-magnetic-resonance-image-analysis

Automated cardiovascular magnetic resonance image analysis

Last updated:
ID:
83198
Start date:
28 April 2023
Project status:
Closed
Principal investigator:
Professor Hongshan Yu
Lead institution:
Hunan University (HNU), China

The goal of our project is to develop an automatic and fast segmentation method for cardiac images. We plan to spend 36 months dissecting each part of the heart accurately. After that, the volume and thickness of each part of the heart were measured and counted. This will facilitate the application of computer technology in medical image processing. And it may help guide our health.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/automated-detection-of-ocular-systemic-disease-via-retinal-fundus-imaging

Automated Detection of Ocular & Systemic Disease Via Retinal Fundus Imaging

Last updated:
ID:
17643
Start date:
1 March 2016
Project status:
Closed
Principal investigator:
Mr Philip Nelson
Lead institution:
Google LLC, United States of America

Retinal imaging in the form of fundus photography and OCT are well-established diagnostic tools for eye diseases such as diabetic retinopathy, glaucoma and age-related macular degeneration. It has also been suggested as a prognostic tool for the severity of systemic disease such as diabetes, stroke, and dementia. Our work centers around using machine learning and computer vision to (1) automate the detection of eye diseases which are currently diagnosed via retinal imaging and (2) identify novel features in retinal imaging that may be predictors or early signs of eye disease as well as systemic disease.
If successful, this work will help improve the detection of eye diseases and potentially other systemic diseases. Automated detection also has the potential of increasing efficiency and reducing costs. Using labeled fundus and OCT images as the main inputs, we will train computer algorithms to automatically predict image labels using machine learning and computer vision. We are requesting data from all patients that have had the retinal imaging performed. Per the UK Biobank look-up tool, this consists of 67,711 patients that make up the collection of 68,151 paired colour retinal photographs and optical coherence tomography (OCT) scans.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/automated-detection-of-sleep-apnea-via-predictive-model-based-on-patient-health-and-medical-data

Automated detection of sleep apnea via predictive model based on patient health and medical data

Last updated:
ID:
48716
Start date:
19 June 2020
Project status:
Current
Principal investigator:
Dr Niranjan Sridhar
Lead institution:
Verily Life Sciences LLC, United States of America

Our goal is to be able to extract predictive factors to construct a sleep apnea diagnosis predictive model for identifying people that are at high risk of sleep apnea and co-morbidities associated with sleep apnea. Research has shown that majority of the US population alone has about 80% undiagnosed OSA individuals. Our screening diagnosis algorithm using this predictive model based on health and medical data hopes to reach undiagnosed OSA patients at scale to reduce patient burden of clinical path of diagnosis for those that are at high risk of OSA. Identified high risk OSA individuals will be offered the opportunity to be educated about OSA to long-term treatment adherence, in order to improve patient sleep quality in relation to clinical outcomes, especially in co-morbid and high risk and high-cost populations. For examples, cardiovascular and metabolic diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/automated-disease-and-disease-risk-detection-from-retinal-images-with-deep-learning

Automated disease and disease risk detection from retinal images with deep learning

Last updated:
ID:
45682
Start date:
25 March 2019
Project status:
Closed
Principal investigator:
Mr Bart Elen
Lead institution:
VITO, Belgium

The retina offers a unique window on a person’s health. The smallest blood vessels, present everywhere in our body, are visible in the eye and not covered by skin. This allows to detect small changes to the blood vessels which are indicative of increased risks for cardiovascular, neurological and systemic diseases. Furthermore, pictures from the retina can be taken in a quick and non-invasive way, giving it a high potential as screening tool.
In this 3 year project we want to study how artificial intelligence can be applied to automatically detect diseases and increased risks for diseases from retinal images. This research has the potential to have a large impact on public health. In the future, it could allow to act on upcoming diseases before quality of life is reduced. Enabling us to increase the number of healthy life years we can enjoy while reducing public healthcare expenses.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/automated-feature-based-detection-grading-and-staging-of-sight-threatening-eye-conditions-in-colour-fundus-photographs

Automated Feature-Based Detection, Grading, and Staging of Sight-Threatening Eye Conditions In Colour Fundus Photographs

Last updated:
ID:
74127
Start date:
30 November 2021
Project status:
Current
Principal investigator:
Dr Nicolas Jaccard
Lead institution:
Project Orbis International, Inc., United States of America

Aims:
The aim of this project is to develop an automated system for the detection of sight-threatening eye conditions. This system will allow a machine to interpret eye images the same way a clinician would. The type of image this system will read is called Fundus photographs, which are images of the back of the eye that are used to assess eye health. Fundus photographs are used at multiple level of cares, from optometrists to specialised secondary care settings.
Crucially, this system will not simply provide a decision such as “disease detected” or “no disease detected”, but will instead provide a granular report that can be directly related to existing diagnosis procedures. This way, the system can be used to supplement the decisions made by the healthcare professional, rather than overriding them.

Scientific Rationale:
Globally, 253 million people are blind or visually impaired. This disproportionately impacts patients in low- to middle-income countries (LMIC).
Artificial Intelligence (AI), which allows computers to be trained to carry out specific tasks, has been hailed as one of the solutions to alleviate the effects of this public health emergency. However, it has yet to demonstrate its full potential, especially in low- to middle-income countries (LMIC) where it is likely to be the most impactful. This is due to economic factors, lack of evidence based on patient data originating from these regions, and products that focus on overriding the user’s decisions rather than supplementing them.
We want to take advantage of the UK Biobank data to build a system that will be truly useful when used on-the-ground. Based on previous experience trialling AI approaches in multiple clinical settings, we identified an approach that is likely to be successful in doing so. Instead of relying on computer algorithms that are not informative for the healthcare professional users (so-called “black box” system), we opted to develop a system that would provide granular information in a way that is directly relatable to diagnosis procedures the user is familiar with.

Project Duration:
The project is estimated to last up to three years.

Public Health Impact:
We expect that this project is likely to have a significant impact on the diagnosis and treatment of those conditions in LMIC. Imaging date annotations to be shared back will likely spur more research in this area.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/automated-generation-of-musculoskeletal-phenotypes-from-the-uk-biobank-extended-imaging-study-augment-study

AUtomated Generation of Musculoskeletal phENotypes from the UK biobank exTended imaging study (AUGMENT Study)

Last updated:
ID:
17295
Start date:
1 August 2016
Project status:
Current
Principal investigator:
Dr Ben Faber
Lead institution:
University of Bristol, Great Britain

We aim to further develop and refine a suite of automated analyses generating secondary variables from DXA scans. These methods will then be used to derive a comprehensive set of musculoskeletal phenotypes from 100,000 participants in the extended imaging study, including hip and knee shape, vertebral fractures and scoliosis. The relationship between these DXA phenotypes and clinical outcomes related to osteoporosis and osteoarthritis, such as fractures and joint replacements, will subsequently be explored. We also plan to identify novel molecular pathways involved in the pathogenesis of musculoskeletal disease, based on genetic factors associated with DXA phenotypes and clinical outcomes. We aim to build a major musculoskeletal research resource for Biobank, by using state-of-the-art DXA methods, and deliver an augmented musculoskeletal phenotype for future Biobank researchers.

We then aim to use these newly derived data to better understand the determinants of musculoskeletal disease, for example spinal fractures (as seen on DXA VFA scans), joint shape (hip and knee – which may predispose to osteoathritis), scoliosis (which can cause back pain). These musculoskeletal diseases are common and therefore our findings will have important impact for health throughout our society. Whole body scans:
a. Regional bone density will be extracted using semi-automated methods to check for artefacts and regional alignment
b. Scoliosis (curvature of the spine) will be derived based on automated methods in development

Hip scans:
a. Principal components of hip shape will be obtained using automated methods in development
b. Hip structural analysis will be performed using available automated software to extract measures of hip strength

Knee scans:
Principal components of knee shape will be obtained using automated methods in development

Whole spine scans:
Vertebral fractures/ deformities will be detected using automated methods after further developmental refinement Phase 1: (n=5000) methods development
Phase II: (n=100,000) methods application
(i.e. the full cohort in whom DXA is available)


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/automated-grading-and-risk-prediction-of-diabetic-retinopathy-using-uk-biobank-retinal-fundus-images

Automated Grading and Risk Prediction of Diabetic Retinopathy Using UK Biobank Retinal Fundus Images

Last updated:
ID:
996855
Start date:
16 September 2025
Project status:
Current
Principal investigator:
Mr Zhenwei Ye
Lead institution:
First Affiliated Hospital of Jinan University, China

Research Questions:

Can UK Biobank fundus image data be effectively used for the automatic typing of diabetic retinopathy (DR)?

Is it possible to construct an accurate DR risk prediction model based on these image features and clinical data?

What is the performance of the proposed model in predicting the onset and progression of DR?

Objectives:

To develop a deep learning model capable of analyzing fundus images from the UK Biobank database and automatically identifying and classifying diabetic retinopathy.

To integrate multi-modal data, including fundus image features, genetic information, lifestyle factors, and clinical indicators, to construct a comprehensive risk prediction model for diabetic retinopathy.

To validate and evaluate the accuracy and clinical utility of the constructed model for predicting the onset and progression of DR.

Scientific Rationale for the research:
Diabetic retinopathy is one of the leading causes of blindness worldwide. Early diagnosis and risk prediction are crucial for effective disease management. Traditional diagnostic methods rely on subjective assessments by ophthalmologists, which are often inefficient and subject to variability. The UK Biobank, with its vast and diverse dataset of fundus images, genetic and clinical data, provides a unique opportunity to develop and validate innovative, AI-based tools for DR diagnosis and risk prediction. This study aims to leverage this data to build an objective and efficient automated system. Such a system would improve early screening, risk stratification, and management of DR, thereby reducing the burden on healthcare systems and ultimately improving patient outcomes.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/automated-macular-thickness-measurements-and-association-with-potential-risk-factors-and-systemic-disease

Automated macular thickness measurements and association with potential risk factors and systemic disease

Last updated:
ID:
2112
Start date:
4 March 2013
Project status:
Current
Principal investigator:
Mr Praveen Patel
Lead institution:
Moorfields Eye Hospital NHS Foundation Trust, Great Britain

Optical coherence tomography imaging (OCT) rapidly produces 3 dimensional images of the macula (the sensitive part of the retina used for central vision). Abnormalities of macular thickness and structure on OCT imaging are the hallmark of both diabetic retinopathy (commonest cause of vision loss in working aged individuals in the UK) and age-related macular degeneration (commonest cause of vision loss in the elderly). OCT imaging can also provide information about the thickness of the macular nerve fibre layer, which may be thinned in glaucoma (commonest cause of irreversible vision loss worldwide) and in neurodegenerative diseases.
Our proposed analysis falls into 3 components: (1) use high speed computer algorithms to generate automated macular thickness measurements from OCT images captured from the 78,880 UKBiobank participants who underwent OCT imaging; (2) subject 5% of the analysed images and images which failed automated analysis to a manual grading process (3) use automated approaches to determine retinal sublayer thicknesses.
We are requesting access to OCT images and data on lifestyle and medical history. This will allow us to report the distribution of macular thickness across the sampled Biobank population and enable us to gain a clearer understanding of factors that influence macular thickness in health and disease.
The proposed analysis is well aligned with UK biobank objectives as deriving macular thickness measurements from the stored OCT data could help improve the diagnosis and treatment of a wide-range of serious and life-threatening diseases including eye diseases (age-related macular degeneration and glaucoma) and systemic disease (diabetes and neurodegenerative diseases).


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/automated-screening-for-paroxysmal-atrial-fibrillation-from-sinus-rhythm-ecgs

Automated screening for paroxysmal atrial fibrillation from sinus rhythm ECGs.

Last updated:
ID:
35452
Start date:
24 April 2018
Project status:
Closed
Principal investigator:
Dr James Alastair Fraser
Lead institution:
University of Cambridge, Great Britain

We aim to develop a low-cost automated screening test for paroxysmal atrial fibrillation (PAF), a common, treatable disease associated with stroke. This will allow detection of PAF sufferers from their sinus rhythm ECGs.
Our proposed research follows from preliminary work in a small clinical study that demonstrated a change in the complexity of sinus rhythm ECGs in PAF sufferers. The UK Biobank has a unique record of resting and exercise ECGs and cardiovascular diagnoses that will now allow us to develop and test an algorithm that is suitable for screening for PAF in the normal population.
Our research aligns with the UK Biobank?s purpose by aiming to improve the diagnosis of a common, serious but underdiagnosed condition.
Paroxysmal atrial fibrillation (PAF) increases the risk of ischaemic stroke roughly five-fold but around half of cases are undiagnosed. Cases are frequently missed because PAF often occurs in short episodes interposed with long periods of normal sinus rhythm, and can be symptomless. Our work aims to prevent potentially life-threatening consequences of PAF by allowing many more sufferers to receive appropriate anti-clotting therapy.
Resting ECGs from the UK Biobank will be analysed using a new algorithm. In a small study, we have shown that this algorithm can detect an abnormal heart rhythm called paroxysmal atrial fibrillation, even if the ECG appears normal. We now need to test this in many more subjects and assess whether other cardiovascular conditions also influence ECG complexity. We will begin by analysing half of the resting ECGs within the UK Biobank (~2500 records) to learn how complexity measures correlate with the cardiovascular diagnoses of the same subjects. We will then blind-test this tool against the remaining ECGs. The study will initially look at all subjects with a resting 12-lead ECG (5082 records). It will then look at a 10,000 patient subset of the exercise ECG records to explore whether exercise ECGs might be more predictive of paroxysmal atrial fibrillation than resting ECGs.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/automated-segmentation-of-brain-white-matter-tract-based-on-dmri

Automated segmentation of brain white matter tract based on dMRI

Last updated:
ID:
91455
Start date:
27 October 2022
Project status:
Current
Principal investigator:
Ms Wan Liu
Lead institution:
Beijing Institute of Technology, China

White matter (WM) is intrinsically related with various cognitive and behavioral functions, and the study of its structure is important in brain research. Diffusion Magnetic Resonance Imaging (dMRI) provides a unique non-invasive imaging method to study white matter. It utilizes the anisotropy of water diffusion in tissues to obtain the directional information of fibers. Furthermore, fiber tracking technology was used to reconstruct brain connections, where nerve fibers are used to represent streamlines.

Since different brain functions involve different brain regions, we can further divide white matter into different types of WM tracts according to the brain regions connected by fibers, so as to obtain specific neural pathways and conduct more specific analysis of WM. The division of WM tracts is also named as WM tract segmentation. WM segmentation can be achieved by classifying the fiber streamlines derived from fiber tracking or by directly labeling the voxels, i.e. volumetric segmentation. It provides an important quantitative tool for brain connectivity analysis and is widely used in the study of brain structure, brain development and brain diseases.

The initial segmentation of WM tracts was achieved by manual delineation. Experts use anatomical knowledge to manually select the three-dimensional streamlines representing nerve fibers obtained based on fiber tracking technology, and obtain the fiber streamlines of interest as the segmentation result of WM tracts. However, the time cost of manual delineation is high and the reproducibility is poor. Therefore, researchers proposed an automated segmentation method to achieve objective and efficient WM tract segmentation. Early automated methods used registration or traditional machine learning algorithms to achieve WM tract segmentation. In view of the excellent performance of deep learning in various image processing tasks, methods based on deep learning are also applied to WM tract segmentation and greatly improve the accuracy of segmentation.

In view of that the segmentation performance of the existing methods on some WM tracts is still limited especially when only a small amount of labeled data is available, this research aims to propose the innovative method for WM tract segmentation based on dMRI and deep learning to further improve the accuracy of WM tract segmentation and further promote the application of WM tract segmentation in brain disease research. Our project duration is at least 3 years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/automated-segmentation-of-visceral-organs-and-tissues-in-whole-body-mri

Automated segmentation of visceral organs and tissues in whole body MRI.

Last updated:
ID:
40040
Start date:
18 June 2019
Project status:
Closed
Principal investigator:
Dr Sergios Gatidis
Lead institution:
University of Tübingen, Germany

In this research project we want to build a program that can find organs automatically in medical images. In the UK Biobank imaging study, thousands of volunteers are examined using Magnetic Resonance Imaging (MRI). The resulting data can give insights into processes within the human body. However, it would take too much time to analyze all the data manually. Thus, an automated analysis program is necessary.
In our project we will use machine learning methods in order to create a tool for automated detection and delineation of organs on MRI. In order to do so, we will train a machine learning algorithm by presenting data that were already analyzed by humans. As a next step, this algorithm can learn to fulfill the respective task automatically.
We hope that this project will contribute to a faster and better analysis of MRI data in the context of the UK Biobank MR study.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/automatic-and-trustworthy-radiomics-for-cardiovascular-diseases

Automatic and Trustworthy Radiomics for Cardiovascular Diseases

Last updated:
ID:
100972
Start date:
10 November 2023
Project status:
Current
Principal investigator:
Ms Fanwen Wang
Lead institution:
Imperial College London, Great Britain

Aim:
The project aims at building an automatic model for trustworthy radiomics feature extraction and correlate it with a specific disease. The numerical quantifiers of shape and tissue characters extracted from the model can be combined with qualitative descriptors to facilitate more promising and reproducible clinical reports.

Scientific rationale:
The images we acquired under the clinical settings are not just pictures but data including information about disease-specific processes. However, the cardiac and respiratory motion makes CMR-based radiomics extremely challenging. By incorporating novel registration and segmentation, we intend to develop novel algorithms to extract, analyse and model many medical features correlated with specific diseases, improving medical decision-making.

Project Duration:
36 months

Public Health Impact:
With advanced registration and segmentation ability, the proposed methods will extract novel imaging biomarkers with biological context with informative radiomics. We hope the proposed model will enjoy strong reproducibility and explainability, hence having the potential to be translated into the clinical setting.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/automatic-assessment-and-quantification-of-cardiovascular-mri-exams

Automatic Assessment and Quantification of Cardiovascular MRI exams

Last updated:
ID:
30769
Start date:
12 December 2018
Project status:
Current
Principal investigator:
Dr Puneet Sharma
Lead institution:
Siemens Medical Solutions USA, Inc, United States of America

In this study, we will develop machine learning based algorithms for automatic assessment and quantification of cardiac MRI exams in the UK Biobank cohort. Automated image processing is a key enabler in facilitating biomarker discovery in large cohorts such as the one in UK Biobank. Results from this automated analysis will be made available to the research community for further statistical analysis jointly with other data collected for the same subjects. We will develop computer algorithms that will automatically analyze the individual images from the entire cardiac MRI exam for each subject and identify, characterize and quantify key anatomical entities (such as the chambers of the heart, the motion of heart muscle, flow in the aorta etc.). The algorithms will derive measurements from these images in a standardized manner, obviating the need for tedious manual work. We will include the entire Cardiac MRI cohort (100,000 subjects) in this study.In addition to the imaging data, we also request some other metadata such as age, sex, height, weight, body mass index, heart rate, systolic and diastolic blood pressure, and 12-lead ECG data at rest, smoking history, medication status, results of the blood test (cardiac specific biomarkers).


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/automatic-diabetic-retinopathy-detection-based-on-deep-learning-using-retinal-images-and-clinical-data

Automatic Diabetic Retinopathy Detection based on Deep Learning Using Retinal Images and Clinical Data

Last updated:
ID:
46758
Start date:
14 May 2019
Project status:
Current
Principal investigator:
Dr Rodrigo Varejao Andreao
Lead institution:
Federal Institute of Espirito Santo, Brazil

Diabetes mellitus (DM) is an important and growing health problem for all countries, regardless of their degree of development. In 2015, the International Diabetes Federation (IDF) estimated that 8.8% (95% confidence interval [CI]: 7.2 to 11.4) of the world population aged 20-79 years (415 million people) lived with diabetes. If current trends persist, the number of people with diabetes is projected to exceed 642 million by 2040. One consequence of DM is diabetic retinopathy (DR). Such disease affects vision and is the leading cause of vision loss. However, the early stage of the DR is asymptomatic and when detected it could be late. Early diagnosis and control of the disease are very important to reduce the damage caused by diabetic retinopathy.

Many studies have shown diabetic retinopathy detection systems based on retinal image classification alone. However, in a more realistic setting, the use of clinical data can be very relevant both to improve the accuracy of diagnostic aid systems, to help the screening of such diseases at the Primary Health Care and to understand the evolution of the disease at early stage.

High-performance diabetic retinopathy detection systems could help the screening of such disease even though a specialized opinion is not present which is the case at the Primary Health Care. As a consequence, the prognosis of the disease will improve as well as the effect of the treatment.

The objective of this project is to investigate whether automatic diabetic retinopathy detection can benefit from the usage of both retinal images through its features extracted by deep learning algorithms and clinical data such as weight, height, waist circumference, blood glucose, glycated hemoglobin, and others.

The project should be completed in 36 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/autophagy-related-causes-of-cardiovascular-disease

Autophagy-related Causes of Cardiovascular Disease

Last updated:
ID:
57535
Start date:
24 June 2020
Project status:
Current
Principal investigator:
Dr Rajeev Malhotra
Lead institution:
Massachusetts General Hospital, United States of America

Vascular disease is the leading cause of disability and death in the UK and worldwide. Autophagy, the cell-based process in which cells degrade potentially dangerous compounds and waste products, has a potential a role in the prevention of vascular diseases. When cells are prevented from properly disposing of dangerous compounds or cellular waste products, the risk for vascular disease and related illnesses increases. We seek to use the UK Biobank to identify individuals with vascular disease and individuals free from vascular disease. Comparing the genetic profiles of these two groups can help identify components of the autophagy pathway that relate to vascular disease. We will further explore findings from these comparisons by investigating how genetic mutations in genes related to autophagy contribute to the risk of vascular disease using cultured cells and mice models. Bringing insights from the population level to the research bench is a powerful strategy to gain understanding of the biology of vascular disease and identify useful targets for disease prevention and treatment. We anticipate that analysis of the dataset will take nine to twelve months with ongoing investigation in our laboratory.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/based-on-multi-omics-data-to-explore-the-impact-of-factors-in-terms-of-lifestyle-environmental-risk-treatment-and-genetic-characteristics-on-the-pathogenesis-and-prognosis-of-cardiovascular-diseases

Based on multi-omics data to explore the impact of factors in terms of lifestyle, environmental risk, treatment and genetic characteristics on the pathogenesis and prognosis of cardiovascular diseases

Last updated:
ID:
545415
Start date:
27 May 2025
Project status:
Current
Principal investigator:
Dr Hao Cui
Lead institution:
Beijing Anzhen Hospital, Capital Medical University, China

Cardiovascular diseases (CVDs) are the leading cause of death worldwide, which is a serious concern especially in aging society because age is one of the most important risk factors for CVDs. CVD burden is determined by diverse conditions, ranging from hypertension and ischemic heart disease to persisting infection-related diseases that affect the economy and health systems. The etiology of CVDs is multifactorial, with among lifestyle factors, treatment strategies, genetic characteristics playing critical roles. Previous studies have shown that lifestyle behaviors and environmental factors, including physical activity, diet, BMI, and environmental pollution are associated with CVD and long-term diseases.Besides, specific genetic mutations are responsible for CVDs.

Although several mechanisms have been proposed, the molecular dysregulations linking lifestyle factors or genetic characteristics and CVDs are still unclear. Plasma metabolome and proteome provide a comprehensive set of circulating metabolites and proteins. Besides, Radiomics can also provide clinical or preclinical biomarkers from medical images. Whether these factors are associated with CVDs and to what extent they mediate lifestyle behaviors or genetic characteristics and CVDs remain unclear.

It is important to clarify the impact of these elements on the pathogenesis, treatment and prognosis for the effectively prevention of CVDs and other long-term diseases. The main purpose of this study is to determine specific factors in terms of lifestyle, environmental risk, treatment strategies and genetic characteristics elevate the risk of CVDs, based on multi-omics data. We will first explore the impact of lifestyle, environmental factors, genetic characteristics and treatment strategies on the incidence and mortality of CVDs and long-term diseases. On this basis, we will further study the characteristics of metabolomics, proteomics and radiomics, and their association with the development of diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/based-on-multimodal-data-to-explore-the-associations-between-lifes-eight-essentials-and-hydration-status-and-cardiometabolic-multimorbidity-and-other-age-related-diseases-in-older-adults

Based on multimodal data to explore the associations between life’s eight essentials and hydration status and cardiometabolic multimorbidity and other age-related diseases in older adults

Last updated:
ID:
98698
Start date:
22 November 2023
Project status:
Current
Principal investigator:
Dr Yinqiao Dong
Lead institution:
Shanghai Jiao Tong University School of Medicine., China

Aims: This study intend to focus on the older adults (aged 50 years old) to explore the associations between life’s 8 essentials, hydration status and cardiometabolic diseases and other aged-related diseases (including cardiovascular disease, chronic kidney diseases, osteoporosis, cognitive function, sarcopenia and mental health).
Scientific rational: The trend of global population aging is increasing due to increased longevity. Recently, GBD 2019 data found that ischemic heart disease, stroke, and diabetes remain the leading causes of death in the global elderly population. Healthier lifestyles such as stopping smoking, increasing physical activity and having a balanced diet (e.g., eating more fruit and vegetables and reducing salt intake) have been broadly acknowledged as critical factors in reducing the burden of diseases. In addition, proper hydration is also critical to physiological functioning and health. The impact of low water intake and unhealthy lifestyle on health is under-researched using UK biobank. Therefore, it is crucial to explore the impact of low water intake and an unhealthy lifestyle (based on the Life’s 8 Essentials proposed by the American Heart Association) on cardiometabolic diseases and other aged-related chronic diseases, especially in the elderly population, as this could affect many people in the UK. Existing studies might be limited by small sample size, suboptimal control for important confounders, or both. Therefore, we proposed to investigate the associations between life’s 8 essentials, hydration status and cardiometabolic diseases and other aged-related diseases and this study based on the genetic data, biochemistry markers, the use of medications and imaging feature using genetic risk score (GRS) and deep learning methods to further reveal the potential mechanisms.
Project duration: This project is part of a PhD thesis expected to last 3 years (36 months), considering the time for data clean, statistical analysis, manuscript writing and sending the manuscript to a medical journal with possible revisions.This project will be extended if the research cannot be completed on time.
Public health impact: This study will provide new directions and epidemiological evidence for middle-aged and older adults to prevent cardiometabolic diseases and other aged-related diseases. If increasing water intake and adherence to a healthy lifestyle has the potential to reduce the risk of these health outcomes, it is important to conduct research using the UK Biobank and inform the public accordingly. Informing the public is critical, especially because current large sample population-based cohort studies of the effects of dehydration and lifestyle are inadequate.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/baseline-observational-study-of-the-similarities-and-discordances-in-biomarkers-and-lifestyle-factors-determining-lifespan-between-european-north-american-and-japanese-populations

Baseline observational study of the similarities and discordances in biomarkers and lifestyle factors determining lifespan between European, North American and Japanese populations.

Last updated:
ID:
82557
Start date:
7 March 2022
Project status:
Closed
Principal investigator:
Dr Matt Glasgow
Lead institution:
Edanz Group Japan Inc., Japan

There are significant differences between European, North American, and Japanese populations. These have practical implications in a wide range of areas from the design of clinical treatments and pharmaceuticals to how lifestyle factors can be modified to produce better health outcomes.

This research aims to address what are the main differences between Japanese, European and North American populations with respect to physical and biochemical measures, disease progression, and lifespan.

The research aims to identify how population-based biases in factors relate to lifespan. Further the research aims to identify generalised predictors of lifespan, and develop methods for relating Japanese, North American, and European population data for future genomic association studies.

The rationale for the study is to undertake a comprehensive outcome driven study to understand the associations of phenotypic markers, including biometric and biochemical markers, and diagnostic-intervention combinations, with risk of death.

This study will seek to characterise the dominance of each marker’s association with lifespan and how these associations affect an overall lifespan predictor. This will provide clear data for how to relate different population data to lifespan while accounting for biases within each sample.

A better understanding of the differential associations between clinical and biomedical data and health outcomes within European, North American, and Japanese population datasets will be of growing importance for large population studies looking to understand genomic and polygenic links to disease. There is also a public health need to understand ethnicity-driven bias and consider better people’s characteristics and cultural/lifestyle factors.

The project will last up to three years in duration.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/bayesian-models-for-gene-and-behavioral-interaction-on-cancer-and-cardiovascular-morbidity-and-mortality

Bayesian Models for Gene and Behavioral Interaction on Cancer and Cardiovascular Morbidity and Mortality

Last updated:
ID:
44804
Start date:
26 March 2019
Project status:
Closed
Principal investigator:
Dr Yelena N Tarasenko
Lead institution:
Georgia Southern University, United States of America

Many studies have documented main or independent effects of various demographic, lifestyle, genomic, clinical, and psychosocial factors on cancer and cardiovascular outcomes and engagement in physical activity. However, we know little about interaction effects; i.e., the combined effects of multiple exposures or risk factors and effects of one exposure within strata of another. An improved understanding of these effects is essential for uncovering synergistic or differential relationships and preventing development of ineffective or mistargeted interventions. Hence, our goal is to identify interaction and effect modification within the physical activity and cancer control framework.

Because interactions are rare and usually unknown, identifying them from data has proven to be challenging, in terms of statistical inference and the requisite computation in potentially high-dimensional spaces. Our first aim is to develop novel Bayesian approach which will offer several advantages over an already scant number of approaches to detecting interactions. Our second aim is to apply the developed methodology to the UK Biobank – “the world’s largest objective physical activity dataset now available,” which “redefined what is possible in the field of physical activity epidemiology,” and paved the way to “robustly examine associations and interactions between activity, diseases, environmental factors and genetics” (UK Biobank, 2/2/2017).

The project will be executed in three years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/bayesian-statistical-methods-for-air-pollution-and-mental-health-research-in-a-changing-climate

Bayesian Statistical Methods for Air Pollution and Mental Health Research in a changing climate

Last updated:
ID:
86234
Start date:
9 September 2022
Project status:
Current
Principal investigator:
Ms Abi Isabella Riley
Lead institution:
Imperial College London, Great Britain

The main aim of this project is to describe the long-term effect of air pollution on mental health outcomes in the UK. It is expected to be a three-year project, under PhD funding from the Medical Research Council.
Objectives:
1. Assimilate numerical models with ground-level monitoring station and satellite-derived measurements for air pollution and climate, to create a new air pollution exposure model for the UK.
2. Use machine learning algorithms to classify land use in the UK for a measure of proximity to greenspaces and typology.
3. Develop a model for quantifying the effects to air pollution on mental health outcomes, while investigating if nearby greenspaces could alleviate these potential negative effects.
4. Perform causal inference between long-term exposure to air pollution and mental health.
The current global focus on the climate crisis and air pollution has brought new developments in GIS methods, satellite imagery, and open source data. Similarly, the area of mental health research has rapidly gained interest from a variety of stakeholders, including international governments, research organisations, and individuals, as seen in the recent UN Change Conference. Therefore, this project positioned in the intersection between health, environment and biostatistical research fields, will provide unique insights on the relationship between polluted environment and mental health.
Underpinning this project are a number of methodological advances too. Bayesian statistics is a current significant area of interest in mathematics, especially its innate ability to incorporate multiple data sources and updated data. Similarly, the use of Big Data and machine learning are both growing fields in industry and academia. This project will take advantage of large, open-source datasets and will use machine learning for characterizing greenspaces from satellite imagery. Furthermore, causal inference is a really hot methodological topic; many current studies on air pollution and mental health just focus on correlation, even so ‘correlation does not imply causation’.
Finally, by including location, physical health, and socioeconomic variables within the models, we can identify trends in mental health outcomes across the country and in different demographic and vulnerable groups. By identifying these groups, mental health awareness campaigns, treatment resources, and medical training can be distributed appropriately. Similarly, any identified trends in the effects of air pollution, climate, and greenspaces on mental health can help to form more specific targets in the battles to reduce emissions, combat climate change, and promote the development and use of greenspaces and parks for leisure and exercise.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/behavioral-environmental-and-genetically-related-predictors-of-biological-aging-and-aging-related-diseases

Behavioral, environmental, and genetically related predictors of biological aging and aging-related diseases

Last updated:
ID:
233885
Start date:
3 January 2025
Project status:
Current
Principal investigator:
Professor Ying Shao
Lead institution:
First Hospital of Jilin University, China

As our global population ages, tackling biological aging and treating diseases linked to aging, like diabetes, Alzheimer’s disease, and heart disease, are urgent priorities. These conditions not only affect the quality of life for millions but also put a heavy strain on healthcare systems worldwide. Our study seeks to uncover patterns and outcomes of these age-related diseases, identify what factors contribute to their development, and understand how we can effectively delay aging itself.
Aims:
(1) Understanding Disease Patterns: We aim to map out how age-related diseases develop and spread across different groups of people. By understanding these patterns, we can identify who is most at risk.
(2) Assessing Disease Impact: We will evaluate how these diseases affect people’s health, quality of life, and longevity. This will help us create better treatment and prevention plans.
(3) Identifying Predictive Factors: Our research will pinpoint the key factors that lead to these diseases, especially those we can change, like lifestyle choices and environmental factors. This knowledge will guide us in developing strategies to prevent these conditions.
(4) Exploring Molecular Mechanisms: We also plan to dive deep into the molecular mechanisms that contribute to biological aging and these diseases. By understanding these processes, we aim to find new targets for interventions that could extend the healthy years of life.
Methodology:
Our research will use a combination of traditional statistical methods and modern data mining techniques to analyze a comprehensive dataset, which includes genetic, lifestyle, environmental, and biochemical information. This approach allows us to create predictive models that can identify high-risk individuals and suggest targeted prevention strategies.
Duration: The project is expected to span over three years.
Public Health Impact:
The insights gained from our research will have significant public health benefits. By uncovering the pathways that delay aging and identifying effective interventions for age-related diseases, we aim to reduce the disease burden, improve healthcare outcomes, and enhance the quality of life for the elderly population. This will not only benefit individuals but also lead to more efficient resource allocation in healthcare systems.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/bidirectional-association-between-cardiovascular-disease-and-prostate-cancer-in-a-prospective-cohort-study

Bidirectional Association Between Cardiovascular Disease and Prostate Cancer in a Prospective Cohort Study

Last updated:
ID:
332912
Start date:
15 October 2024
Project status:
Current
Principal investigator:
Professor Jianye Wang
Lead institution:
Beijing Hospital, China

Scientific rationale: Over the past few decades, the burden of morbidity and mortality from cardiovascular disease (CVD) and prostate cancer has been steadily increasing. Given the clinical co-occurrence of CVD and prostate cancer in observational studies, it is reasonable to believe that a recursive relationship may exist between these two diseases.

Major Objective: We aimed to evaluate bidirectional associations between CVD and prostate cancer. Moreover, we assessed whether this bidirectional association differs across genetic risk levels. In addition, we applied MR analysis to evaluate the potential causal relations of two conditions.

Project duration: 3 years

Public health implications: Investigating the recursive relationship between CVD and prostate cancer is important to facilitate the personalized prevention of both diseases. Further insights into cardiac oncology may lead to targeted intervention and treatment for individuals with cancer.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/bidirectional-associations-between-somatic-conditions-and-neurodevelopmental-disorders-a-cohort-study-based-on-the-uk-biobank

Bidirectional associations between somatic conditions and neurodevelopmental disorders: A cohort study based on the UK Biobank.

Last updated:
ID:
676416
Start date:
27 May 2025
Project status:
Current
Principal investigator:
Professor Zhixiong Liu
Lead institution:
Xiangya Hospital of Central South University, China

Our research aims to investigate the bidirectional relationships between somatic conditions and neurodevelopmental disorders, as well as the impact of neurodevelopmental disorders on patient mortality. Specifically, we will examine how various somatic conditions-such as hypertensive disorders, dyslipidemia, autoimmune diseases (e.g., rheumatoid arthritis, celiac disease), metabolic diseases (e.g., diabetes, obesity), cardiovascular diseases, cancer, and psychiatric disorders-contribute to the development of neurodevelopmental disorders, primarily autism spectrum disorder (ASD), attention deficit hyperactivity disorder (ADHD), and intellectual disability (ID). Conversely, we will also explore whether these neurodevelopmental disorders increase the risk of developing somatic conditions and assess their potential influence on patient mortality.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/big-data-analyses-of-the-long-term-benefits-of-knee-articular-cartilage-surgery

Big data analyses of the long-term benefits of knee articular cartilage surgery

Last updated:
ID:
280752
Start date:
5 November 2024
Project status:
Current
Principal investigator:
Dr Nicola Kuiper
Lead institution:
Keele University, Great Britain

Does knee cartilage repair surgery prevent arthritis or the need for a joint replacement?
In the UK, approximately 1/5 of over-45s have been diagnosed with osteoarthritis (OA), the knee being the most frequently affected joint. The cause of OA is unknown, but it develops when cartilage wears out. For example, knee injury in young adults can increase their risk of developing knee OA sixfold. There is also a genetic contribution to OA. OA is treated with physiotherapy, painkillers, surgery, or as a last resort, a joint replacement. Cartilage repair surgeries focus on stopping knee pain and improving function, but they may also slow OA. It’s important to measure outcomes following surgery to understand if they have helped the patient. Ideally, knee cartilage surgeries in middle-aged patients will slow down OA and delay or eliminate the need for joint replacement. We don’t know if this happens in real-time. We plan to work out the true effect of knee cartilage surgeries by studying health records from the UK Biobank to find out whether cartilage surgery can help patients with a higher genetic risk of OA onset. We will bring together several risk factors (e.g. patient characteristics, other disorders, cartilage surgery type and genetic risk score for OA) to create a tool (a phone app) for orthopaedic surgeons. They will use the tool in clinics to help fit cartilage treatment to their patients to delay or avoid the development of severe OA. It will help weigh up the benefits of surgery relative to risk. This will improve their quality of life and reduce the need for knee replacements, both of which represent health-economic benefits.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/big-data-and-artificial-intelligence-in-cardio-metabolic-disease

Big Data and Artificial Intelligence in Cardio-Metabolic Disease

Last updated:
ID:
148816
Start date:
19 November 2024
Project status:
Current
Principal investigator:
Dr Jie Shi
Lead institution:
University of Science and Technology of China, China

Scientific rationale: Cardio-metabolic disorders, encompassing a spectrum of conditions from heart diseases to metabolic dysfunctions, pose significant challenges in modern healthcare. Often silently progressing, these disorders can evade early detection, amplifying the subsequent health implications and socio-economic burdens. The emergence of advanced omics technologies has heralded a new era in precision medicine, providing novel insights into the molecular foundations of diseases.

Aims: Our research seeks to revolutionize the landscape of early diagnosis and intervention. By harnessing advanced technologies and vast data sources, complemented by the power of artificial intelligence, we aim to gain a deeper understanding of these disorders. Integrating diverse streams of health data – from lifestyle nuances and genetic blueprints to proteomic and metabolic fingerprints, complemented by medical imaging – we aspire to construct a comprehensive tableau of cardio-metabolic health.

Project duration: The duration of the project will be for 36 months.

Public health impact: The cornerstone of our approach is the application of AI. AI’s computational prowess will synthesize this multi-modal data, enhancing our capability to unearth subtle early markers and predict potential disease trajectories. Such AI-driven insights promise not only precision but also timeliness in intervention, two critical facets in managing such complex disorders. Our endeavor, rooted in this innovative confluence of multi-omics and AI, holds the promise to transform the paradigm of cardio-metabolic healthcare. By facilitating a deeper, more holistic understanding of disease mechanisms, we envision a future where personalized, proactive interventions become the norm, significantly mitigating the health and economic impacts of these disorders. In essence, our research stands at the crossroads of advanced technology and clinical expertise, aspiring to shape the future of cardio-metabolic healthcare.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/big-data-based-analysis-of-genotype-phenotype-correlations-in-eye-diseases

Big data-based analysis of genotype-phenotype correlations in eye diseases

Last updated:
ID:
87083
Start date:
24 May 2022
Project status:
Current
Principal investigator:
Dr Zhenzhen Liu
Lead institution:
Zhongshan Ophthalmic Center, SYSU, China

Eye disease is a growing public health concern worldwide. Eye disease-related blindness and visual impairment carry a significant global economic burden. For example, the annual cost of lost global productivity due to vision impairment from uncorrected myopia and presbyopia alone is estimated at $244 billion and $25.4 billion, respectively. Different eye diseases require different and timely management. The identification of specific environmental influences and genetic influences that affect eye diseases will facilitate the implementation of effective interventions. This will improve the health care of eye disease and reduce the public health burden associated with eye disease. All work is expected to be completed within the next 36 months, but it may be extended due to methodological advances and new discoveries that may require external validation. This project is conducive to more effective prevention and resolution of eye problems in patients with eye diseases, to avoid visual impairment and reduce the burden of disease caused by eye diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/big-data-driven-evidence-generation-to-advance-personalized-therapeutic-strategies-for-chronic-metabolic-diseases-through-genetic-clinical-and-pharmacological-insights

Big Data-Driven Evidence Generation to Advance Personalized Therapeutic Strategies for Chronic Metabolic Diseases Through Genetic, Clinical, and Pharmacological Insights

Last updated:
ID:
604474
Start date:
5 April 2025
Project status:
Current
Principal investigator:
Professor In-Wha Kim
Lead institution:
Seoul National University, Korea (South)

Research Aims:
This study aims to advance the treatment of chronic metabolic diseases by generating evidence for pharmacological strategies and developing personalized therapies. By leveraging genetic data, we will predict drug effects, validate these findings through cohort studies using real-world data, and synthesize robust evidence. Additionally, by exploring genetic factors that influence individual drug responses, we seek to establish personalized therapeutic strategies tailored to genetic profiles.

Scientific Rationale:
Drugs often influence multiple pathways, affecting various clinical outcomes. Genetically mimicked drug target variations can provide insights into their effects on chronic metabolic disease prognosis. Large-scale real-world data enables transparent, reproducible evidence generation. Understanding pharmacogenetic factors behind individual drug response variability supports personalized treatment strategies.

Methods:
We will conduct three main types of analyses:
1. Genetic data from the UK Biobank will support Genetic Risk Score (GRS) to estimate the impact of drug targets on disease prognosis.
2. Cohort Studies: We will create prospective and retrospective cohorts to compare drug users and non-users using UK Biobank and NHS-linked data. Korean real-world data will validate findings, and meta-analysis will synthesize results.
3. Pharmacogenetic Analysis: A cohort of drug users will be analyzed to identify genetic variations (e.g., star alleles). Differences in clinical outcomes will be evaluated based on these variations, identifying genetic factors that contribute to variability in drug response.

Public Health Impact:
This research combines genetic epidemiology and real-world data to predict therapeutic effects, validate drug effectiveness, and explore genetic factors influencing drug responses. These findings will support personalized treatment strategies, improving care and outcomes for chronic metabolic disease patients.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/big-data-driven-sleep-genes-identification-in-human

Big data driven sleep genes identification in Human

Last updated:
ID:
62001
Start date:
30 March 2021
Project status:
Current
Principal investigator:
Dr Koji L Ode
Lead institution:
University of Tokyo, Japan

Humans sleep for about eight hours. On the other hand, it has been reported that other animals, such as koalas, sleep for about 16 hours. Since the duration of sleep differs from organism to organism, it is expected that there is a gene that determines the duration of sleep, and a major goal of recent sleep research is to find the gene that determines the duration of sleep (sleep gene). In this study, we will look for sleep genes by analyzing activity data and genome data in participants of UKBiobank with short or long sleep duration.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/big-data-for-mobility-research

Big Data for Mobility Research

Last updated:
ID:
27871
Start date:
27 March 2018
Project status:
Closed
Principal investigator:
Dr Scott Delp
Lead institution:
Stanford University, United States of America

We are interested in determining how intrinsic (age, gender, diseases) and extrinsic (e.g., public transit access) factors affect physical activity. For example, in a recent study based on step-count data from 2 million individuals, we found that unique measures of physical activity predict BMI and sleep quality. The UK Biobank accelerometry data would help us validate and extend these findings. After characterizing a baseline for activity in individuals without musculoskeletal limitations, we would like to understand how osteoarthritis (OA), one of most common disorders that limit mobility, and its treatment (joint replacement surgery), affect activity patterns. Our Mobilize Center is an NIH-funded Big Data to Knowledge Center of Excellence that is using advanced data science methods to improve the current understanding of conditions that limit mobility. The proposed research should meet the UK Biobank?s stated purpose by facilitating the development of new interventions aiming to restore mobility patterns that are beneficial to human health. Previous studies that have analyzed functional outcomes at different stages of osteoarthritis (OA) or after joint replacement surgery are limited to in-lab tests for small sample sizes. Using activity-monitoring data from the UK Biobank, we will study the debilitating effects of OA and the hypothesized improvement after joint replacement surgery at a large scale and in free-living settings. For comparison, we will also characterize activity patterns in individuals without musculoskeletal limitations. All the subjects who participated in the activity-monitoring segment of the study. (If possible, also all the subjects who have OA-related symptoms or have had joint replacement surgery.)


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/big-data-integration-methods-and-applications-to-uk-biobank-data

Big Data Integration Methods and Applications to UK Biobank Data

Last updated:
ID:
19039
Start date:
11 January 2017
Project status:
Closed
Principal investigator:
Professor Hulin Wu
Lead institution:
University of Texas (UT Health), United States of America

Aim 1: Develop novel high-dimensional modeling approaches to integrate genotyping, phenotype and biomarker data for the prediction of death and cancer outcomes.
Aim 2: Develop novel network approaches to integrate genotyping and multimodal imaging data (including MRI and DXA scans) to predict death and cancer outcomes.
Aim 3: Develop novel time course modeling approaches to model accelerometry, dietary and behavioral data to predict death and cancer outcomes. Raw accelerometer data will allow us to understand the true signal and the noises, which is important for the statistical methodology. We expect that our new approaches will result in important health science findings from the UK Biobank data, since we expect to integrate genotype, biomarker, dietary, behavioral and imaging data (including MRI and DXA) to predict death and cancer outcomes. We will follow the UK Biobank policy to share our methodologies and research results with the general public.
We intend to use imaging data (including MRI and DXA) to define possible novel variables and also to verify or derive new approaches for data pre-processing. Imaging data will be used since these are potential predictors of chronic diseases and cancer. We will employ big data approaches for the preparation and analysis of the data provided by the UK Biobank. These approaches will be based on standard statistical methods and techniques. Which methods are used in each approach can only be determined from the characteristics of available data.
This project?s output includes novel statistical and computational approaches to integrate the variety of complex data in the UK Biobank for outcome predictions. From the use of these novel approaches we expect to obtain novel predictions of diagnostics of death and cancer. Full Cohort


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/big-data-mining-and-deep-learning-to-improve-risk-prediction-early-diagnosis-and-prognosis-evaluation-of-chronic-diseases

Big Data Mining and Deep Learning to Improve Risk Prediction, Early diagnosis and Prognosis Evaluation of Chronic Diseases

Last updated:
ID:
106528
Start date:
7 September 2023
Project status:
Current
Principal investigator:
Dr Na Shen
Lead institution:
Tongji Hospital, China

Chronic diseases including cancer, diabetes and cardiovascular diseases are the leading causes of disability and death in the world, imposing a huge economic and disease burden to the society. It is well-known that chronic diseases are the result of long-term combination of genetic predisposition, environmental factors, physiological conditions and behavior patterns, which also affect the prognosis or health-related outcomes of chronic diseases. Previous studies and our team have made some efforts on risk factors, early diagnosis or prognosis of chronic diseases, but results are often different and warrant further investigation in large cohorts such as the UK Biobank.

Multidimensional data mining and deep learning approaches have shown quite excellent performance in risk prediction, early diagnosis and prognosis evaluation of many diseases. This project will integrate comprehensive data, such as genetic, environmental, behavioral factors, laboratory indicators, imaging data and omics data, to discover potential risk factors contributing to onset and development of chronic diseases, and fit optimal models to show which factors could improve risk prediction, be potential biomarkers for early diagnosis or survival assessment, and what mechanisms may explain these effects. By state-to-art deep learning techniques, we can also identify high-risk populations and those who may benefit from screening or monitoring.

We plan to conduct a three-year study by using data available from the UK Biobank. This study will provide novel models to improve risk prediction, early diagnosis and prognosis evaluation of chronic diseases, which have important implications for health management of chronic diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/biobank-behavioural-biomarkers-b4m

BioBank Behavioural BioMarkers (B4M)

Last updated:
ID:
21770
Start date:
11 May 2018
Project status:
Closed
Principal investigator:
Professor Aldo A. Faisal
Lead institution:
Imperial College London, Great Britain

Ageing is an important risk factor in many common diseases and is also associated with observable differences in posture and behaviour. Behaviour provides a rich phenotype that can be collected at a largescale, and is affected by a wide range of underlying physiological changes, making it a useful integrative phenotype. However, identifying which aspects of this complex time varying phenotype are most useful remains a challenge. We aim to use machine learning to identify optimal combinations of parameters that will serve as behavioural biomarkers of ageing and disease by taking advantage of the crosssectional data available in the UK biobank. Our purpose is develop data-driven behavioural biomarkers of ageing and disease that will eventually enable us to provide behaviour-driven diagnostics, monitoring and stratification of diseases and ageing. Behavioural biomarkers are especially appealing as they can be collected using wearable sensors, such as existing smartphones and smartwatches and therefore offer a large-scale low-cost opportunity to promote health throughout society. Thus, our aims align perfectly with UK Biobank project?s goals of improving diagnosis of illness and promoting health throughout society. We will perform analysis of movement/behavioural data in the UK Biobank and employ pattern recognition algorithms to extract meaningful features of behaviour (behavioural fingerprinting). We will then correlate these features with age and disease related variables and use pattern classification techniques to develop polymarkers of age and disease based on behavioural motion patterns.
We aim to to analyse the full cohort for which high-resolution accelerometer data are available (currently listed on the website as 103,711 participants). This is because we expect from our own experience to date that Biobank behaviour data is highly variable depending on context and individual differences. We aim to develop an objective marker that is defined


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/biobank-brain-and-cardiac-mutual-risk-indexing-bbc-mri-study

Biobank Brain and Cardiac Mutual Risk Indexing (BBC MRI) study

Last updated:
ID:
18545
Start date:
1 April 2016
Project status:
Current
Principal investigator:
Professor Paul M Matthews
Lead institution:
Imperial College London, Great Britain

The prevalence of both brain and cardiac disease rises with age and there are functional interactions between the two organs in health and disease (the heart-brain axis).  However, cardiac and brain co-morbidities in later life are poorly explored and risk factors or markers that may suggest interacting pathophysiological mechanisms remain to be elucidated.  The aim of our research is to investigate brain-heart axis using brain and cardiac MRI images and carotid IMT data in the context of clinical, genetic and lifestyle (e.g., smoking, alcohol use) data in UK Biobank. The results of our study will lead to better understanding of brain-heart axis and may improve the prevention, diagnosis and treatment of both brain and cardiac diseases in later life. We will perform analyses of the heart and brain images and review clinical histories of people in UK Biobank to identify signs of diseases such as stroke or signs of impaired heart or brain functions. We will work to understand relationships between these and how they may be influenced by a person?s genes or other medical conditions or their lifestyle. We hope to understand how to better assess the risk of brain and cardiac disease in later life and how the health of the two organs is related. To have the greatest power for the range of nested analyses, data from the full cohort of subjects with imaging data available at the time of this application are requested (~5000 individuals anticipated). As future imaging data are able to be released, we would like to supplement the study group to test exploratory hypotheses generated from the initial (~5000 subject) data using a test dataset covering an additional 15,000 people.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/biochemical-and-genetic-architecture-of-transfusion-and-transplantation

Biochemical and genetic architecture of transfusion and transplantation

Last updated:
ID:
74245
Start date:
8 September 2021
Project status:
Current
Principal investigator:
Dr Mikko Arvas
Lead institution:
Finnish Red Cross Blood Service, Finland

Blood transfusion is a common treatment for blood loss in severe trauma and elective surgery. Organ transplantations are required to replace failed organs while hematopoietic stem cell transplantation replaces the recipient’s immune system. All of these are lifesaving treatments and they can be commonly referred to as tissue transplantation. Tissue transplantation is costly as it requires advanced infrastructure to guarantee the safety of both tissue donors and recipients. Tissue transplantation is also ethically complex as it depends on living or deceased volunteer donors. Hence, ensuring and improving efficacy of tissue transplantation has a large societal impact.

Population scale biochemical and genetic data from projects such as UKBB and FinnGen allows for unprecedented deciphering of biochemical and immunogenetic architecture of tissue transplantation. What is the impact of genetics on who is most likely to benefit from a transplantation or suffer adverse effects from donating tissues? Can available blood assay data provide us with biomarkers or mechanistic insight for the above topics? Genetic tissue matching has been used for decades improve success of tissue transplantation, but how does the variation outside conventional blood group and histocompatibility genes impact on success of tissue transplantation? What are the genetic factors behind immune-related traits? Our project will bring light to these crucial topics during the next 3 years by combining genetic and clinical data over several different condition-specific smaller cohorts to UKBB and FinnGen projects population scale data.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/biochemical-estimation-of-muscle-mass-and-unbiased-assessment-of-kidney-function-using-serum-creatinine-and-cystatin-c-a-prognostic-validation-study-within-uk-biobank

Biochemical estimation of muscle mass and unbiased assessment of kidney function using serum creatinine and cystatin-C. A prognostic validation study within UK Biobank.

Last updated:
ID:
102574
Start date:
2 June 2023
Project status:
Current
Principal investigator:
Dr John Richard Prowle
Lead institution:
Queen Mary University of London, Great Britain

Multimorbidity is a word used to describe a person with two or more long-term health conditions. It is a growing problem for healthcare as we are living longer. This means we are more likely to develop long-term health conditions.

Kidney disease is an important long-term condition linked to heart disease. Heart disease is a leading cause of death. Patients with kidney disease need more hospital appointments and blood tests to stay healthy. They are more likely to be admitted to hospital with a serious illness than a healthy person. One of the main jobs of your kidney is to filter waste and toxins. A blood marker called creatinine is used to diagnose kidney disease. It is used to estimate how well your kidney is filtering waste and toxins. Muscle creates most of creatinine.

As you get older, losing some of the muscle you have is normal. It also does not work as well as it used to. When you are sick this happens faster than usual. This process is sarcopenia. We know that people who have sarcopenia when they are sick are more likely to die. They are also less likely to recover well from their illness.

People who are sick will make less creatinine if they have sarcopenia. This means that diagnosing people with kidney disease when they are unwell may not be accurate. Cystatin-C is another blood marker of kidney function. It is not affected by the amount of muscle you have. It can be used to diagnose people with kidney disease. It may be a better marker of kidney disease when people are unwell.

We will explore the link between kidney disease, muscle mass, creatinine and cystatin-C. We will do this using the UK Biobank data.

We aim to answer the following questions:
– Can this link predict patient outcomes after admission to hospital? This will include death or the development of kidney and heart conditions.
– Can we use cystatin-C to better predict the development of kidney disease after someone is unwell?

We expect this project to take 3 years to complete. Our goal is to improve the identification of kidney disease in patients with critical illness. This means that we will be able to prevent and treat more kidney disease. This will have a positive impact on multimorbidity.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/biochemical-evaluation-of-secondary-osteoporosis-a-uk-biobank-study

Biochemical evaluation of secondary osteoporosis: A UK Biobank study

Last updated:
ID:
23448
Start date:
20 March 2018
Project status:
Closed
Principal investigator:
Fadil Hannan
Lead institution:
University of Liverpool, Great Britain

Osteoporosis is a common disorder characterised by reduced bone strength and an increased susceptibility to broken bones. Osteoporosis may occur secondary to other potentially treatable medical conditions, and the study aim is to determine the usefulness of blood tests for diagnosing these secondary causes of osteoporosis. This study will be undertaken by comparing whether subjects with osteoporosis, as diagnosed by the presence of low bone mineral density (BMD) values and/or fractures, have more frequent blood test abnormalities compared to subjects without osteoporosis. These findings will establish which blood tests are of help for the clinical assessment of osteoporosis. Osteoporosis represents a global health burden, and in the UK causes more than 300,000 fractures every year, with annual hospital costs of greater than £1.9 billion (National Osteoporosis Society). The goal of this study is to identify laboratory tests that can be used to diagnose osteoporosis caused by potentially treatable medical conditions such as hypogonadism, vitamin D deficiency and primary hyperparathyroidism. This objective is therefore in keeping with the aims of the UK Biobank, which are to improve ?the prevention, diagnosis and treatment of a wide range of serious and life-threatening illnesses ? including?osteoporosis?? (UK Biobank website). We propose to evaluate the frequency of abnormal osteoporosis related biomarkers, which are currently included in the UK Biobank panel (e.g. serum calcium, alkaline phosphatase, creatinine, vitamin D, and testosterone) to assess whether alterations in these biomarkers occur more commonly in the following participant groups:
1. Participants with low impact fractures compared to participants without low impact fractures.
2. Participants from the DXA-assessment cohort who have osteoporotic bone mineral density (BMD) values compared to participants with osteopaenic (i.e. mildy reduced) or normal BMD values.
We propose to evaluate the following participants:
1. Participants who provided information on experiencing a fracture after a simple fall (>49,000 participants; data-field 3005)
2. DXA-assessment cohort participants who have had spine, hip and whole-body BMD measured (~10,000 participants; category 125)

For our primary analysis, ‘osteoporosis’ is defined as a femoral neck BMD T-score of =-2.5, based on NHANES III gender-specific data. We will define ‘severe osteoporosis’ as low BMD and the presence of a low trauma fracture since age 40. These definitions are in line with the WHO osteoporosis working group.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/biochemical-signatures-of-lean-mass-and-biological-age

Biochemical signatures of lean mass and biological age

Last updated:
ID:
70960
Start date:
17 May 2021
Project status:
Current
Principal investigator:
Dr Thomas R Wood
Lead institution:
University of Washington, United States of America

Slowing the rate of ageing and reducing age-related chronic disease (such as heart disease and dementia) is perhaps the most important public health mission of the 21st century. In order to do this we need better ways to track how fast somebody is ageing before they develop significant disease. PhenoAge is a metric of biological age, rather than chronological age, which reproducibly and strongly predicts both disease risk and risk of death. However, another important predictor of longevity is activity, strength, and muscle mass. Some of the tests used to predict increased biological age with PhenoAge also increase in those who exercise more or have more muscle mass, so this project aims to see whether having more muscle makes you look “older” on PhenoAge when in fact it should do the opposite – reduce the risk of death and chronic disease. If that is the case, the research will provide updated predictions that are more accurate after taking into account how much muscle mass somebody has. This will be useful to both the public tracking their own health and doctors, scientists and public health officials who may implement tracking of biological age in response to drug or lifestyle interventions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/bioimpedance-defined-lean-tissue-mass-in-chronic-kidney-disease-its-relationship-to-comorbidity-mortality-and-surrogate-markers-of-frailty

Bioimpedance defined lean tissue mass in chronic kidney disease: its relationship to comorbidity, mortality and surrogate markers of frailty.

Last updated:
ID:
70918
Start date:
30 November 2021
Project status:
Current
Principal investigator:
Dr Matthew Tabinor
Lead institution:
Keele University, Great Britain

Many long-term health conditions lead to progressive loss of muscle bulk, usually referred to as lean tissue mass (LTM). This loss of muscle bulk can be particularly severe in people with chronic kidney disease, contributing to the most important symptom experienced by these patients: fatigue. Bioimpedance is an established method which safely estimates the muscle bulk in a patient at the bedside. Questions remain, however, as to whether there is value in measuring muscle bulk routinely and how it should be interpreted in clinical practice. We principally still do not know: (1) “how much is loss of muscle bulk explained by reduced kidney function compared to the effects of other long term health conditions?” and (2) “To what extent does the loss of muscle bulk explain how likely it is for patients to die, suffer from adverse events (such as falls) and symptoms attributable to frailty, such as fatigue, in patients with chronic kidney disease?”.

To investigate these questions we will use data collected by the UK Biobank, a long-term study of half a million people of whom 50,000 have one or more long-term health condition. Muscle bulk was measured using bioimpedance when patients entered the study. What happened to these patients was subsequently recorded throughout the study. We will use a novel statistical approach to understand how long-term health conditions and the level of kidney function in turn affect muscle bulk in patients with different demographic characteristics. This will enable us to describe whether these factors act on a patients muscle bulk, and how this relates to their symptom burden, health-related outcomes and the risk of death.

Our research findings will inform clinicians how best to interpret bioimpedance data measuring reduced muscle bulk by understanding: (1) how loss of muscle bulk is influenced by other long-term health conditions and (2) the relative importance of losses in muscle bulk in the development of adverse events and symptoms. It is likely this research will reinforce the need to take a more personalised approach to clinical decision making where treatment goals may be influenced by the presence of other long-term health conditions. Our findings will be disseminated to patients and other relevant stakeholders, multidisciplinary conferences to healthcare professionals who work with patients with multiple long term health conditions, including chronic kidney disease, and by publication in peer reviewed scientific journals.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/bioinformatics-approaches-that-integrate-multi-omics-data-to-identify-genetic-risk-factors-for-complex-diseases

Bioinformatics approaches that integrate multi-omics data to identify genetic risk factors for complex diseases.

Last updated:
ID:
774758
Start date:
30 July 2025
Project status:
Current
Principal investigator:
Professor Zhijie Han
Lead institution:
Chongqing Medical University, China

Complex diseases, including cancer, neurodegenerative disorders, and cardiovascular diseases, are among the leading global health challenges. Although GWAS studies have successfully identified numerous genetic markers associated with these diseases, their underlying pathogenic mechanisms remain largely unclear. Recent advances in single-cell and spatial transcriptomics have enabled a deeper exploration of disease mechanisms by revealing cellular heterogeneity, gene expression dynamics, and spatially resolved regulatory networks within the tissue microenvironment. The integration of these technologies offers a more comprehensive and in-depth perspective for precision medicine. However, the integration of highly heterogeneous and large-scale multi-omics datasets remains challenging, particularly across cohorts with varying sample sizes, emphasizing the need for innovative analysis strategies.
To advance our understanding of the genetic architecture of complex diseases and their interactions with environmental factors, we propose to integrate genomic data with multi-omics datasets, including metabolomics, proteomics, environmental exposure, and both single-cell and spatial transcriptomics data. The UK Biobank, with its extensive population-scale dataset and rich genetic, clinical, and environmental information, presents an unparalleled opportunity for such integrative analyses. We aim to leverage UK Biobank data alongside external multi-omics datasets to identify novel disease-associated genetic variants and their corresponding risk factors, validating our findings across independent cohorts. To achieve this, we will develop and refine bioinformatics tools to enhance the efficiency of data integration and analysis, enabling a systematic exploration of genetic risk factors, gene regulatory mechanisms, and environmental influences on disease susceptibility. This project offers valuable insights that will advance precision medicine and personalized treatment strategies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/bioinformatics-methods-for-precision-cardiovascular-medicine

Bioinformatics methods for precision cardiovascular medicine

Last updated:
ID:
50978
Start date:
30 October 2019
Project status:
Closed
Principal investigator:
Dr Jason H Moore
Lead institution:
University of Pennsylvania, United States of America

A central goal of precision medicine is to tailor disease risk assessment, diagnosis, and treatment to specific biochemical, cellular, clinical, demographic, environmental, and genomic characteristics of individual patients or sub-groups from the population for improving health and healthcare. However, there are significant challenges related to developing precision medicine strategies from population-based results, since statistical summaries derived from a human population do not explicitly provide information about the health of an individual. The overarching goal of this 3-year project is the development of bioinformatics methods and software for connecting population-based models of heart failure susceptibility with individual-level measures to advance cardiovascular precision medicine. This will result in a Virtual Genomic Medicine workbench (VGMed) to enable experimentation and hypothesis generation relative to cardiovascular medicine, accelerating the translation of genomic findings relative to heart failure into the clinic.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/bioinformatics-methods-for-the-genetic-analysis-of-infectious-diseases

Bioinformatics methods for the genetic analysis of infectious diseases

Last updated:
ID:
51190
Start date:
30 October 2019
Project status:
Closed
Principal investigator:
Dr Jason H Moore
Lead institution:
University of Pennsylvania, United States of America

An important goal of infectious disease research is to develop genetic predictors of susceptibility. The overarching goal of this 3-year project is thinking about and approaching the genetic analysis of infectious disease from a complex systems point of view. Genetic variants include common and rare variants and both are expected to play an important role in common diseases. In contrast to common variants, rare variants are difficult to analyze because there are so few individuals in the population that have the rare allele. This makes it difficult to statistically compare cases and controls. One answer to this has been to collapse multiple rare variants across a genomic region, but the current approaches are simplistic in that they treat all variants as being equal and additive while at the same time ignoring their spatial context. We will develop a biologically-inspired approach to the analysis of rare variant associations methodology that can identify optimal subsets of rare variants and, at the same time, identify the optimal way to collapse them into new common variants that can be used in genetic association studies. We will then analyze combinations of the resulting collapsed variants together with actual common variants for their association with infectious diseases, such as HIV/AIDS and tuberculosis. This will be done via non-parametric and genetic model-free machine learning approaches that will enable us to embrace the complexity of the relationship between genomic variation and phenotypic variation.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/biological-age-unveiled-a-drop-of-blood-as-the-key-to-predicting-the-aging-clock-with-machine-learning

Biological Age Unveiled: A Drop of Blood as the Key to Predicting the Aging Clock with Machine Learning

Last updated:
ID:
797219
Start date:
26 June 2025
Project status:
Current
Principal investigator:
Dr Yibo Wang
Lead institution:
Xinqiao Hospital of Army Medical University, China

Aim: Develop a comprehensive aging clock model using multi-omics data from the UK Biobank, including clinical, proteomic, and metabolomic datasets. This model based on machine learning will predict physiological age and organ senescence, and assess disease risk from a single blood test.
Research Questions:1. How can multi-omics data be integrated to accurately forecast an individual’s biological age and organ aging? 2. What is the model’s accuracy and generalizability across different databases and independent clinical sample repositories? 3. How can the model be optimized and simplified for practical medical application through a minimally invasive blood test?
Objectives:1. Construct an aging clock model based on integrated multi-omics datasets. 2. Validate the model’s predictive accuracy in external datasets and clinical samples. 3. Refine the model for practical use in clinical settings with the finger-blood test technology we developed.
Scientific Rationale:Emerging evidence suggests that multi-omics data provide a holistic view of the aging process, capturing changes in gene expression, protein function, and metabolic pathways. By analyzing these datasets, we aim to identify biomarkers of aging that can be used to assess an individual’s physiological age and organ health. And we are already advancing low-cost technology for detecting target biochemical substances in fingerstick blood tests, which will support the technical feasibility of our model.. Our ultimate goal is to create a user-friendly, clinically applicable model that requires only a drop of blood, offering insights into organ senescence and disease risk prediction.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/biological-and-environmental-determinants-of-pancreas-size-and-their-association-with-diabetes

Biological and Environmental Determinants of Pancreas Size and Their Association With Diabetes

Last updated:
ID:
79758
Start date:
8 February 2022
Project status:
Current
Principal investigator:
Professor Jack Virostko
Lead institution:
University of Texas (UT Austin), United States of America

We want to find out why people have different sized pancreases and whether having a small pancreas increases someone’s risk for getting diabetes. We will identify relationships between someone’s pancreas size and their diet, early life experiences, and health history. This will help us understand what a ‘normal’ pancreas size is for each person. By knowing a ‘normal’ pancreas size for each person, we can find out when someone’s pancreas size is smaller than we expect, which may mean they are at risk of getting diabetes. We will find out whether there are differences in the relationship between pancreas size and other measurements in individuals with and without diabetes. This retrospective project will be completed in 2 years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/biological-correlates-of-brain-aging-in-healthy-adults-and-adults-at-risk-for-alzheimers-disease-and-related-dementias

Biological correlates of brain aging in healthy adults and adults at risk for Alzheimer’s disease and related dementias

Last updated:
ID:
343478
Start date:
29 July 2025
Project status:
Current
Principal investigator:
Dr Mohammad Fili
Lead institution:
Oklahoma State University, United States of America

For our study, we seek to: 1) determine what biological changes affect people with Alzheimer’s disease and other dementias; and 2) look at different kinds of brain imaging to uncover which people have normally aging brains vs. abnormally aging brains. For scientific rationale, Alzheimer’s disease and related dementias affect hundreds of millions of people worldwide. By 2050, there will be substantially more people with dementias that impact not only the quality of their life, but have immensely negative impact on caregivers, family, and friends. We are interested in looking at all major systems in the body that are known to contribute to dementia risk or directly impact the brain. We believe in letting the science tell us which aspects of biology can best inform us about who over time has a normally aging versus abnormally aging brain. In using this data, our project will last a minimum of 36 months. We may ask to extend this data use depending on project needs. For public health impact, we believe having more biological signatures of what happens in normally vs. abnormally aging brains will unlock how to slow or stop brain changes that can strongly impact quality of life and independence.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/biology-and-the-tendency-to-engage-in-entrepreneurship

Biology and the tendency to engage in entrepreneurship

Last updated:
ID:
40174
Start date:
18 September 2019
Project status:
Closed
Principal investigator:
Mr Ahmed Maged Nofal
Lead institution:
University of Warwick, Great Britain

In recent years, researchers have begun to uncover various health and biological consequences of business activities. For instance, researchers have shown that shift work induces elevated levels of cortisol and increased risk of chronic diseases (Manenschijn et al., 2011, Ha and Park, 2005, Karlson et al., 2006, Touitou et al., 1990). Despite the various studies that have suggested that entrepreneurship is favourable for countries’ economies (Obschonka et al., 2014, Wolfe and Patel, 2017a), entrepreneurship can also have impact on individuals’ health. For example, studies have found a positive association between entrepreneurship and anxiety and depression (Hessels et al., 2018). Accordingly, further studies that look “at physiological reactions to work environments and organizational reward mechanisms” are needed (Shane, 2009, p.69). This would provide insights into how work environment can be adapted to minimise any adverse effects on individuals’ health and allow people to adjust themselves to the work environments that suits their mental and physical well-being. As part of the project, we also seek to examine the genetic underpinnings and the neural correlates of entrepreneurship to have a better understanding of the mechanisms relating entrepreneurship to individuals’ health and well-being.
This research should take from one year to two years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/biomarker-discovery-in-neurodegenerative-diseases-using-integrated-longitudinal-data-analysis

Biomarker discovery in neurodegenerative diseases using integrated longitudinal data analysis

Last updated:
ID:
341475
Start date:
25 April 2025
Project status:
Current
Principal investigator:
Dr Bhupat H Desai
Lead institution:
NeuroDiscovery AI Inc, United States of America

At NeuroDiscovery AI, we are dedicated to advancing the understanding and treatment of neurodegenerative diseases like Parkinson’s disease, Alzheimer’s disease, and Frontotemporal Dementia, which progressively damage brain cells. These conditions are influenced by a complex interplay of genetics, environment, diet, and lifestyle. As the global population ages, the socio-economic burden needs to be addressed.
Our approach involves a strategic integration of our proprietary health data with the extensive resources from the UK Biobank (UKB), which provides health records, genetic information, and lifestyle data from a large cohort of individuals. This collaboration enhances our capacity to explore the intricate dynamics of neurodegenerative diseases.
Our research tackles three key questions to improve our understanding and treatment of neurodegenerative diseases:
Finding Reliable Disease Markers: We’re searching for specific genes, proteins, or pathways that indicate the onset and progression of these diseases. By analyzing large datasets of biological and clinical information, we aim to discover biomarkers that help diagnose early and guide treatment development.
Grouping Patients for Better Care: People with the same disease can vary greatly. Using advanced tools and data from diverse populations, we aim to identify subgroups of patients based on their unique molecular, clinical, and demographic profiles. This will pave the way for personalized diagnosis and treatment strategies.
Understanding Lifestyle Effects: We’re studying how genetics, diet, lifestyle, and environment combine to influence disease risk and progression. This will help uncover new ways to prevent or manage these diseases.
The methodologies we employ involve cutting-edge computational and statistical tools. This includes statistical analyses to identify links between potential biomarkers and disease symptoms, as well as machine learning techniques to uncover patterns and predict disease trajectories.
The impact of our research could significantly alter public health outcomes. Enhanced capabilities for early detection and monitoring of neurodegenerative diseases can lead to more effective early interventions, potentially slowing disease progression and improving patient quality of life. Insights into how genetic factors interact with lifestyle choices could also guide public health policies aimed at preventing these diseases. Ultimately, our research aims to foster the development of new treatments and improve the health outcomes for individuals affected by these debilitating conditions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/biomarker-profiling-by-nmr-metabolomics-for-the-study-of-chronic-disease-risk-and-underlying-risk-factors

Biomarker profiling by NMR metabolomics for the study of chronic disease risk and underlying risk factors

Last updated:
ID:
30418
Start date:
6 August 2018
Project status:
Current
Principal investigator:
Dr Peter Wurtz
Lead institution:
Nightingale Health Ltd, Finland

Biomarkers measured from blood samples are indicative of the risk for many chronic diseases, such as heart disease and diabetes. We intend to measure blood samples from the entire UK Biobank using a novel technology developed by Nightingale Health Ltd that captures >200 biomarker measures, such as lipids and amino acids, from each blood sample. Data analyses will assess how well these biomarker measures can predict future risk for disease onset. The analyses further aim to clarify the molecular roles of the biomarkers in chronic diseases and underlying risk factors, and clarify genetic and lifestyle contributions to the biomarker levels. The results may improve the ability to predict disease onset, which would allow better targeting of prevention efforts. The detailed metabolic profiling also provides an enhanced understanding of the molecular mechanisms leading to onset and progression of chronic diseases, and may hereby identify causal biomarkers and targets for drug treatment. The resource of >200 metabolic biomarker measures will also benefit numerous other research projects, such as studies of molecular intermediates of diet and other lifestyle factors as well as the examinations of the genetic basis of metabolism. We will measure the blood samples by Nightingale Health?s proprietary NMR metabolomics platform in Finland. The resulting metabolic biomarker measures will subsequently be analysed statistically for association with disease events (prevalence and incidence of all ICD-10 disease categories) and health factors using so-called phenome-wide association approaches covering subclinical disease markers, other blood biomarkers, life style and dietary data, as well as complete genomic information. We will request blood samples and participant data for the full cohort (~500,000 samples; 85 ul serum needed) and, if possible, also for one follow-up time point (~20,000 blood samples). The metabolomics measurements of the entire UK Biobank are expected to be completed within 18-24 months from sample arrival to Nightingale?s laboratory.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/biomarker-study-for-the-early-detection-of-cancer

Biomarker study for the early detection of cancer

Last updated:
ID:
818972
Start date:
23 July 2025
Project status:
Current
Principal investigator:
Professor Pei Wang
Lead institution:
Icahn School of Medicine at Mount Sinai, United States of America

This project aims to develop blood-based protein biomarker panels for the early detection of breast and/or other cancer types.
In a recently published study, we have prioritized a panel of candidate breast cancer blood protein biomarkers (PMID: 35767427) using state-of-the-art targeted mass spectrometry methods. To further refine and prioritize biomarker candidates for assay development for clinical usage, we propose to analyze the UK Biobank Pharma Proteomics data, an excellent resource for screening and validating early detection biomarker candidates for cancers with moderate to high incidence rates. Specifically, we will conduct comprehensive statistical analyses to identify/prioritize blood proteins whose abundances were linked to prevalent and/or incident cancer events for breast, prostate, colon, lung, and other cancer types.
This project fully complies with UKB’s AI guidance in fairness, transparency, and accountability. If we develop any proprietary models, they will return derived parameters to UKB and avoid generating outputs that risk re-identification. No participant-level data will be used in any generative AI model or made publicly accessible.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/biomarkers-and-mechanisms-of-cardiovascular-side-effects-caused-by-cox-2-inhibitors

Biomarkers and mechanisms of cardiovascular side effects caused by COX-2 inhibitors

Last updated:
ID:
20342
Start date:
10 February 2017
Project status:
Closed
Principal investigator:
Professor Jane Mitchell
Lead institution:
Imperial College London, Great Britain

Nonsteroidal anti-inflammatory drugs (NSAIDs) are amongst the most commonly used medications including for arthritis. NSAIDs can also prevent ~50% of some cancers. However NSAIDs cause cardiovascular adverse events, estimated to result in 30,000-100,000 extra heart attacks and strokes each year in the UK. The concern surrounding NSAID-induced cardiovascular adverse events has led to an arrest in drug development, patient anxiety, cautious prescribing of COX-2 selective drugs and the withdrawal of celecoxib for the prevention of cancer. We aim to identify genetic biomarkers that can predict those susceptible to NSAID induced cardiovascular adverse events. The purpose of UK Biobank is ?to improve the prevention, diagnosis and treatment of a wide range of illnesses and to promote health throughout society?. Our research seeks to identify genetic biomarkers that predict cardiovascular adverse drug reactions in people taking these common anti-inflammatory pain medications. Cardiovascular adverse events are increased by around a third in people taking these drugs which impacts on people with (i) arthritis, (ii) heart disease and, because the medicine can prevent cancer, (iii) people at risk of cancer. In this way, our work is entirely in line with the stated purpose of UK Biobank. We will look for genetic variants that are different between people with arthritis taking NSAIDs that did or did not have heart attacks or strokes. To see if the differences in genotypes that we find are present in everyone that has heart attacks or strokes regardless of if they take these drugs, we will also assess these associations with genetic variants amongst people not taking NSAIDs. When these data sets are combined, we will be able to identify any genetic variants that specifically predict people are susceptible to cardiovascular side effects trigged by NSAID use. We are requesting genotyping data from the full cohort which will be analysed in the following groups:
(i) arthritis patients who report NSAID use who have had an acute myocardial infarction or stroke ~1,000 individuals.
(ii) arthritis patients who report NSAID use and who have NOT had an acute myocardial infarction or stroke ~40,000 individuals.
(iii) patients without arthritis or NSAID use but have had an acute myocardial infarction or stroke ?~9,000 individuals.
(iv) patients without arthritis or NSAID use and who have NOT had an acute myocardial infarction or stroke ~450,000 individuals.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/biomarkers-for-brain-disorders-towards-precision-medicine

Biomarkers for brain disorders: Towards precision medicine

Last updated:
ID:
30091
Start date:
6 April 2018
Project status:
Current
Principal investigator:
Dr Guido van Wingen
Lead institution:
Amsterdam UMC Research BV, Netherlands

Neurological and psychiatric disorders pose a large burden on the individual, their families and our society at large. An increasingly aging population makes it imperative that we detect these crippling disorders at an early stage, giving the subject a greater chance at recovery and reduce the cost of healthcare for our society. Brain disorders are linked to abnormal patterns of brain function and structure.

We will use different machine learning techniques to develop models that can predict disease development on the basis of structural and functional imaging and genetic data, which can serve as biomarkers for brain disorders. Identifying individuals at risk of developing severe brain disorders gives us the best chance to intervene at an early stage of the disorder. This could dramatically increase the chance of the individuals having a better quality-of-life and reduce the negative impact on their, family, friends, health care workers and society. We will analyse the structural brain images from the individuals from the UKBiobank to build a deep learning network. This network will be trained to 1) predict the genetic liabilities for specific psychiatric/brain disorders that will be calculated using the genetic information from these individuals, and 2) the development of diseases during follow-up. We will include all individuals with structural (T1, T2, DTI) and/or functional (resting-state, task-based) datasets along with their genetic information whenever available.
We will also include (non-imaging) data from all other participants from whom genetics is available, in order to create the covariates for further analysis based on better estimates of the population structure.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/biomarkers-for-frailty-and-unsuccessful-aging

Biomarkers for Frailty and Unsuccessful Aging

Last updated:
ID:
63081
Start date:
27 August 2020
Project status:
Current
Principal investigator:
Dr Jonathan Afilalo
Lead institution:
Jewish General Hospital, Canada

One of the main manifestations of unsuccessful aging – frailty – is highly prevalent among older adults, yet it is vastly under-diagnosed. A reason for this under-diagnosis is that testing for frailty in clinical practice requires time-consuming inputs from clinicians and patients. Another reason is that testing is often based on questionnaires that lack accuracy and reliability, or physical performance tests that lack feasibility in the acute care setting. Thus, there is an unmet need to objectively and efficiently measure frailty with minimal human work. We propose to accomplish this by studying biomarkers that are associated with frailty, musculoskeletal aging and loss of muscle mass, cardiovascular aging, and age-related diseases or impairments. Specifically, the objectives of the current study are to develop artificial intelligence (AI) algorithms that can diagnose frailty and predict which patients may be at risk of developing frailty, as well as determine whether these novel frailty scores can be used to predict the risk of adverse health events, hospitalization outcomes, and clinical outcomes. By developing a more individualized measure of frailty that is easier for healthcare providers to assess, the public health impact of this two-year research study can help close current gaps in the diagnosis of frailty and provide a tool that can better guide treatment decisions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/biomarkers-for-late-onset-myasthenia-gravis

Biomarkers for late-onset myasthenia gravis

Last updated:
ID:
189667
Start date:
4 December 2024
Project status:
Current
Principal investigator:
Professor Henry J Kaminski
Lead institution:
George Washington University, United States of America

Diseases in which the body’s immune system attacks itself are increasing across the world with a major reason being the aging population. Both cancers and autoimmune diseases increase with age. Our lab group studies myasthenia gravis (MG). In this disease antibodies attack the nerve-muscle communication point and can produce life-threatening weakness. We were investigating patient samples in order to try find proteins that predict how patients do with treatment and accidentally discovered proteins increased in blood only of patients over 50. This later-onset group already was known to differ in biological measures from younger patients. Our discovery has the potential to define the reasons for this difference. However, we need to confirm that the changes we see are not related only to aging. The UK Biobank has an ideal large data set using the same protein detection method that we used (Olink) and would allow us to verify if the proteins detected are specific to myasthenia gravis changes or not. We expect that if verified we will understand the disease better and thereby develop better treatments focused on the late-onset form of the disease. Our discoveries may be relevant to other autoimmune diseases. Given that the data is already in existence we can complete our analysis in less than 6 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/biomarkers-immunological-signatures-and-hypertension-associated-diseases-causal-inference-using-genetic-and-observational-approaches

Biomarkers, immunological signatures and hypertension-associated diseases – causal inference using genetic and observational approaches

Last updated:
ID:
93156
Start date:
24 May 2023
Project status:
Current
Principal investigator:
Professor Tomasz Guzik
Lead institution:
University of Edinburgh, Great Britain

Elevated blood pressure (BP) remains a major modifiable risk factor for mortality worldwide and leads to development of cardiovascular and cerebrovascular diseases. Variation in various BP indices possesses a widely recognized impact on target organs such as brain or heart. Therefore identification of pathways causally affecting BP that may be pharmacologically targeted is of major importance for public health.
In order to address the above question the current project aims to search for novel biomarkers and immunological signatures that causally affect the level of BP, and related cardiovascular traits such as vascular stiffness or coronary heart disease (CHD). This will be accomplished by using state-of the art methods from the field of epidemiology, and (population) genetics.

Advances in genomic research contributed to identification of major loci involved in pathogenesis of complex diseases such as asthma, hypertension, or CHD. A continuous increment of sample size of Genome Wide Association studies (GWAS) allows for constructing robust genetic risk scores and selection of Instrumental Variables (IVs) for Mendelian Randomization (MR) analyses, which can be further used to test causality between (intermediate) traits and diseases. For example, MR approach have provided evidence that genetically defined eosinophilia is causal for asthma development, while blood lymphocyte count may potentially causally affect development of hypertension and CHD. A similar approach can be applied to molecular biomarkers, such as blood level of proteins or metabolites and immunological signatures that may serve as exposure traits included in potentially causal pathways for traits of interest.

Using MR approach, current project aims to identify biomarkers and immunological signatures that are causal for the level of BP and related clinical traits. Additional observational (both cross-sectional and prospective) studies, linking biomarkers (e.g. OLINK-quantified plasma proteins, biochemical blood marker) or genetic variations to BP level and related cardiovascular traits, will be performed using data derived from the UK Biobank with data on various biomarkers and metabolites already available.

In summary, using state of the art genetic causal inference and epidemiological methods the current, 3-years project aims to identify and characterize molecular biomarkers and immunological signatures that may influence BP level and related traits in humans. Therefore it may contribute to a better understanding of hypertension pathophysiology and creating new treatment opportunities.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/biomarkers-lifestyle-and-body-composition-in-relation-to-healthy-aging

Biomarkers, lifestyle and body composition in relation to healthy aging

Last updated:
ID:
55794
Start date:
4 February 2020
Project status:
Current
Principal investigator:
Professor Xian-Bo Wu
Lead institution:
Southern Medical University, China

Interventions to delay aging and promote healthy ageing are still controversial. Observational studies have revealed that within a homogeneous population sample, there are considerable variations in the extent of disease and functional impairment risk, highlighting a need for valid biomarkers to aid in characterizing the complex aging processes. However, the identification of biomarkers is complicated by the diversity of living situations, lifestyle activities, medical condition or even body fat composition. Hence, there has been no identification of a single biomarker or gold standard tool that can characterize successful or healthy aging.
UK biobank has collected extensive information through questionnaires and physical measurements, as well as biological samples and imaging examinations, which can provide many different types of biomarkers (eg, biochemical, genetic or radiomics) that might shed light on healthy ageing. Moreover, the UK biobank has detailed data on the incidence of various diseases during follow-up.
The present proposal is focusing on exploring the associations of molecular, image-based or DNA-based biomarkers with ageing related health risks (cardiovascular disease, cancer, diabetes, cognitive impairment, etc.). We also plan to examine if the links differ by different lifestyle factors and levels of body composition. The results will have important clinical and public health implications.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/biomarkers-of-neurological-diseases

Biomarkers of neurological diseases

Last updated:
ID:
76921
Start date:
7 March 2022
Project status:
Current
Principal investigator:
Dr Minghao Dong
Lead institution:
Huazhong University of Science and Technology, China

There exist many kinds of biomarkers for neurological diseases to diagnosis, such as tau for AD. Yet, some neurological diseases lack of such biomarkers. In order to get a better diagnose and treatment for patients, more and more biomarkers need to be explored. As is reported, inflammatory factors and cells may play an important role in some neurological disease while their causal relationship is still mysterious, which is valuable to be further dissected. Therefore, We aimed to conduct a Mendelian randomization study to explore the potential causal relationship between our interested traits and neurological diseases, in order to find and validate new biomarkers for neurological diseases. The duration of our project is estimated to be 12 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/biopsychosocial-predictors-of-chronic-pain-following-caesarean-delivery-a-uk-biobank-cohort-study-using-multimodal-data

Biopsychosocial Predictors of Chronic Pain Following Caesarean Delivery: A UK Biobank Cohort Study Using Multimodal Data

Last updated:
ID:
791918
Start date:
18 May 2025
Project status:
Current
Principal investigator:
Dr Sarah Ciechanowicz
Lead institution:
Imperial College London, Great Britain

Chronic postsurgical pain (CPSP) following caesarean delivery affects 15% of patients and is associated with impaired function and reduced maternal quality of life. While acute pain severity, mood, and sleep disruption are implicated, mechanistic understanding remains limited, and no validated risk stratification tools exist. This study will investigate biopsychosocial predictors of CPSP using multimodal data from the UK Biobank, including clinical, psychological, behavioural, imaging, biomarker, and genetic domains.

Women undergoing caesarean delivery will be identified via OPCS-4 codes in Hospital Episode Statistics. The caesarean date will serve as the index event. Predictors will be extracted from the first 12 months postpartum; pain outcomes will be derived from questionnaire data collected after delivery.

Research Question:
Which biopsychosocial risk factors are associated with the development of chronic pain after caesarean delivery?

Objectives:
1. Define a caesarean delivery cohort using hospital records.
2. Extract multimodal data:
* Sociodemographics, age, co-morbidities, BMI, obstetric data
* Acute and chronic pain symptoms, affective symptoms, adverse childhood experiences, cognitive function, energy/fatigue
* Subjective and actigraphy-derived sleep metrics
* Brain imaging markers (e.g., grey matter volumes, connectivity) collected at any timepoint
* Inflammatory and metabolic biomarkers
* Polygenic scores and pain-relevant SNPs
3. Model associations with CPSP.
4. Explore mediation via sleep and pain interference (functional pain metrics).
5. Assess the feasibility of predictive modelling using multimodal data.

Findings will provide mechanistic insights and inform future risk stratification strategies for chronic pain in postpartum populations.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/biostatistics-on-health-and-traditional-chinese-medicine-theories

Biostatistics on health and Traditional Chinese Medicine Theories

Last updated:
ID:
83318
Start date:
5 September 2023
Project status:
Current
Principal investigator:
Professor Dongran Han
Lead institution:
Beijing University of Chinese Medicine, China

In Traditional Chinese Medicine Theory, certain symptoms are correlated. For example, people with watery stools are more likely to have cold feet while People with constipation are the opposite. Through data mining and analysis with scientific methods, it is possible to test Traditional Chinese Medicine Theories with real-world data and reveal how environmental factors and symptoms impact human health outcomes.

We aim to analyze the correlation between environmental factors and personal behavior, health, and success, and how they translate to clinical outcomes. How different symptoms and diseases correlate in the context of Traditional Chinese Medicine Theory.

We anticipate that this project will take 36 months to complete. We plan to publish the results of the analysis to help advance understanding of the correlation between minor symptoms and clinical outcomes. We hope our research will lead to advances in preventive medicine.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/birth-weight-and-cardiometabolic-diseases-in-adulthood

Birth weight and cardiometabolic diseases in adulthood

Last updated:
ID:
57733
Start date:
6 March 2020
Project status:
Closed
Principal investigator:
Dr Ningjian Wang
Lead institution:
Shanghai Ninth People's Hospital, China

The rising prevalence of cardiometabolic diseases has been recognized as a public health problem globally. Observational studies have suggested that low birth weight may increases the risk for CVD and metabolic disease in adulthood. However, we do not have enough evidence to verify whether there have causal associations between low and high birth weight and multiple cardiovascular conditions in large-scale cohorts. Therefore, we intend to study causal effects of low and high birth weight on multiple cardiovascular conditions and metabolic diseases. This project will last for 36 months. Findings from this research may establish causal effect of low and high birth weight on cardiovascular conditions and metabolic diseases. These findings will pave a way for future preventive strategies for cardiovascular conditions and metabolic diseases by modifying factors closely associated with birth weight, such as prenatal nutrition status.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/birth-weight-leukocyte-telomere-length-and-coronary-artery-disease

Birth weight, leukocyte telomere length, and coronary artery disease

Last updated:
ID:
142292
Start date:
22 November 2023
Project status:
Current
Principal investigator:
Dr Xiang Zhou
Lead institution:
Second Affiliated Hospital of Soochow University, China

Numerous studies have shown that birth weight, a key indicator of congenital nutritional status, is inversely related to the risks of coronary artery disease (CAD). A recent Mendelian randomization study suggested that metabolic risk factors might mediate partial CAD risks associated with low birth weight, but most of the underlying mechanisms remain understood. Leukocyte telomere length, a cardiovascular aging biomarker, is also inversely associated with CAD risks. Some studies demonstrated that low birth weight may be associated with longer telomere length. However, an observational study, including over 2000 participants, suggested that there may be no correlation between birth weight and telomere length. Therefore, the association between birth weight and leukocyte telomere length remains undetermined, and it is uncertain whether leukocyte telomere length mediates the association between low birth weight and CAD. This study aims to investigate the role of leukocyte telomere length in the association of birth weight with coronary artery disease using a prospective cohort study. The project duration is 36 months. Our study will help clinicians understand the role of leukocyte telomere length in the association of birth weight with coronary artery disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/bivariate-gwa-analysis-of-nicotine-and-alcohol

Bivariate GWA analysis of nicotine and alcohol

Last updated:
ID:
22529
Start date:
15 September 2016
Project status:
Closed
Principal investigator:
Professor Jacqueline Vink
Lead institution:
Radboud University, Netherlands

Epidemiological studies have highlighted how different substance use behaviours are strongly correlated. Alcohol users are more likely to smoke than nondrinkers and the prevalence of heavy alcohol use in smokers is higher than in nonsmokers. Individually, effects of these substances have profound health implications and dual users are at increased risk, especially for developing different types of cancer. Both twin and genetic-risk prediction studies have shown a common genetic basis for poly-substance use. Nevertheless, only limited efforts to identify these common genetic factors have been undertaken. We propose to systematically investigate these genetic commonalities using UK Biobank data. Investigating overlapping genetic vulnerabilities to alcohol use and smoking is of importance for public health. The type of research we propose is essential to obtain more insight into the nature of individual differences in substance use behaviours and, thus, understanding why some people are more vulnerable to suffer from poly-substance use than others. The use of genetic information to identify people at increased risk for addiction can eventually inform clinical practice and contribute to personalized medicine. Furthermore, by investigating genetic variants associated with both smoking and alcohol, we may identify biological mechanisms underlying addictive behaviours. We will use individuals? genotypes information to run a bivariate genome-wide association analysis (GWA) in order to search for genetic variants underlying co-variance of both phenotypes: tobacco and alcohol use. This has been shown (Liu et al., 2009) to be an effective strategy to detect genes that concurrently influence two or more traits (pleiotropic genes). For sufficient power to run genetic association analyses and obtain accurate SNP effects, we require access to the full (available) cohort.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/blood-pressure-and-bladder-cancer-risk-by-use-of-conventional-analysis-of-main-effects-and-in-interaction-with-the-nat2-genotype-and-by-mendelian-randomization-analysis

Blood pressure and bladder cancer risk by use of conventional analysis of main effects and in interaction with the NAT2 genotype, and by Mendelian randomization analysis

Last updated:
ID:
42410
Start date:
16 October 2018
Project status:
Closed
Principal investigator:
Dr Stanley Teleka
Lead institution:
Lund University, Sweden

Bladder cancer (BP) is common in developed countries. Smoking is the most important risk factor, other established environmental risk factors such as cancer-causing agents from the industry (like the dye industry) and x-rays (special form of light) are rare risk factors. Nearly 30% of individuals that get bladder cancer get it because of specific traits that passed down from their parents. Studies that have examine the complete set of human genes have identified a gene called N-acetyltransferase 2 (NAT2) as one of the most important genes that increases the risk of getting BC.
High blood pressure (BP) is one of the most important risk factor for cardiovascular diseases. Between 30 to 60 percent of BP is caused by genes passed down from parents to children and through a numerous genetic experiments, more than 100 genes affecting BP have been found. BP has been linked to cancer overall and some cancers from specific sites, such as the kidneys and large intestines. In relation to BC, research is rare, but one large study investigating the link between BP and BC found a positive relationship among men. However, that study and most other prior studies had weaknesses, such as not having enough data on blood pressure medication and smoking, two risk factors that may change the true association between BP and BC. Mendelian randomization analysis is special technique of investigating the association between a risk factor and a disease using genes that affect the risk factor of interest. Properly using this technique will allow us to overcome the above mentioned weaknesses that affected prior studies and allow us to test if BP truly causes BC. With this in mind, the first aim of the study is to investigate if BP causes BC and the second aim is to investigate if the relationship between BP and bladder changes depending on the NAT2 gene one inherits from their parents.
This study is the first of it’s kind and is expected to fill some of the knowledge gap in BC biology and how BP is related to BC. This will hopefully lead to better prevention and clinical care for BC.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/blood-pressure-estimation-using-smart-wristbands-and-machine-learning-techniques

Blood Pressure Estimation Using Smart Wristbands and Machine Learning Techniques

Last updated:
ID:
853625
Start date:
4 July 2025
Project status:
Current
Principal investigator:
Miss Melek Sayan
Lead institution:
Kedi Mobil Uygulama Anonim Sirketi, Turkiye

This study aims to estimate blood pressure levels using data collected from wearable devices, combined with health-related variables from the UK Biobank. With the increasing popularity of smart wristbands and their potential for real-time health monitoring, we intend to develop machine learning models that can predict systolic and diastolic blood pressure based on various physical, lifestyle, and biometric data points.
Can we build accurate predictive models for blood pressure using non-invasive wearable data?
How do demographic, lifestyle, and biometric variables influence blood pressure levels in the general population?

Objectives:
1. Integrate smart wristband-derived variables (e.g., heart rate, activity) with UK Biobank data.
2. Build and validate machine learning models (e.g., random forest, gradient boosting) to predict blood pressure.
3. Identify key features that influence blood pressure levels to inform preventive strategies.

Scientific Rationale:
High blood pressure is a leading risk factor for cardiovascular diseases. Early detection and monitoring are critical. By leveraging large-scale population data and wearable technology, this study contributes to the development of proactive, personalized health monitoring systems that can assist both individuals and clinicians in managing blood pressure effectively.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/blood-type-genetic-variation-and-its-relationship-to-covid-19-susceptibility

Blood type genetic variation and its relationship to COVID-19 susceptibility

Last updated:
ID:
65473
Start date:
28 October 2020
Project status:
Current
Principal investigator:
Dr Alex Reiner
Lead institution:
University of Washington, United States of America

Recent studies of COVID-19 patients have discovered a link between a person’s ABO blood type and their susceptibility to COVID-19 infection. Studies show a higher proportion of blood type A in COVID-19 positive patients compared to blood type O, suggesting a person’s blood type can influence how susceptible they are to COVID-19. However, studies have not assessed all ABO blood types. For example, the B blood type hasn’t been investigated in detail and there is a common subtype of the A blood type (A2 subtype) which has not been investigated. Studies have also not investigated the contribution of other blood groups (a total of 33 blood groups are known) to COVID-19 susceptibility, even though other blood groups are known to alter the expression of ABO in a person’s bodily fluids. In this proposal, we plan address these limitations by studying in greater detail an individual ABO blood type information (including ‘subtypes’) and expanding analyses to include all other known blood groups. We will accomplish this objective by using a person’s genetic information to assess their blood type rather than the standard, protein-based test.

Blood group systems are inherited molecules that are encoded in our genetic material. Therefore, it is possible to determine a person’s blood type by studying their genetic information. In fact, using genetic data enables investigation of blood groups for which standard (protein-based) typing reagents are unavailable, including blood groups which alter the presence or absence of ABO in a person’s bodily fluids. Our plan is to assess individual blood type information using the UK BioBank genetic dataset and relate that information to COVID-19 infection. This will provide greater detail regarding a person’s blood type and further clarify how a blood type contributes to differences in COVID-19 susceptibility.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/body-and-adipose-tissue-composition-and-risk-of-obesity-related-cancers

Body and adipose tissue composition and risk of obesity-related cancers

Last updated:
ID:
70902
Start date:
30 March 2021
Project status:
Current
Principal investigator:
Dr Thomas Rohan
Lead institution:
Albert Einstein College of Medicine, United States of America

The aim of the study is to examine the association between adipose tissue composition and the risk of obesity-related cancers. Obesity has been related to risk of several types of cancer. Body mass index (BMI) has been used as surrogate measurement of body fat to estimate obesity-related risk of several types of cancer such as those of the colorectum, liver, pancreas, ovary, endometrium and postmenopausal breast. However, although BMI correlates with adipose tissue mass, it cannot distinguish between lean muscle and fat volume. A growing body of evidence has indicated that adipose tissue, and in particular excessive abdominal fat, affects several metabolic mechanisms and promotes tumorigenesis in the overweight and obese. Visceral fat is more strongly correlated with metabolic changes that are associated with an increased risk of some obesity-related types of cancer. It is unclear, however, whether adipose tissue composition contributes to obesity-related cancer risk. New methodologies, such as magnetic resonance imaging (MRI), provide measures of different types of adipose tissue depots in the body. MRI is costly and not easily available in the clinical setting. This highlights the importance of ongoing UK Biobank multi-modal imaging study, which is the world’s largest study of this kind, designed with the purpose of collecting MRI data from 100,000 individuals already enrolled in the UK Biobank. We propose to use all MRI data on adipose tissue, with a focus on the variables representing fat composition in the abdominal region, already available or that will be collected by the study, and to relate these measures to risk of obesity-related cancer occurrence. We will also take into account other relevant factors, using the extensive information on demographic, medical and lifestyle risk factors available in the UK Biobank study.
The results will provide a unique contribution to the knowledge on the obesity-cancer risk relationship and may help to improve strategies to reduce the risk of certain types of cancer.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/body-composition-bc-studies-collaboration-bcsc-project

Body Composition (BC) Studies Collaboration (BCSC) Project

Last updated:
ID:
70816
Start date:
9 August 2022
Project status:
Current
Principal investigator:
Vivek Prasad
Lead institution:
University College London, Great Britain

Globally, obesity levels have risen and account for 13% of the adult population based on World Health Organization data from 2016. Laterally, malnutrition remains to be a major public health problem that can be prevalent among the underweight, normal weight, overweight and obese individuals. Currently, obesity and underweight diagnostic criteria are primarily based on Body Mass Index (BMI). While BMI measurement is low cost and reduces measurement burden, it does not reflect an individual’s body composition(BC) (that is, fat mass and fat-free mass) because it only accounts for height and weight. Accurate diagnosis of obesity and malnutrition is crucial for individuals to obtain proper treatment therefore, the methods for classifying obesity and underweight should be enhanced. Dual X-ray absorptiometry (DXA) is among the most accurate and convenient direct measures of BC and body fat (BF) distribution.

There are discrepancies in the studies conducted using BMI as the measure of obesity. One example is: Obesity is associated with an increased risk of adverse health outcomes in the general population. However, studies that consisted exclusively of patients with chronic diseases suggest that overweight and obese patients may paradoxically have better outcomes than lean patients. Looking on to the discrepancies caused by the use of BMI, there is a need for a “BMI like” disease risk classification based on BF.

Initial critical analysis using the UK Biobank will investigate the effects of different levels of BC parameters (fat mass, fat-free mass, visceral fat) and changes in these parameters on morbidities, obesity-related mortality and all-cause mortality. Then the UK biobank data will be merged with datasets consisting of DXA measured BC from other countries to construct a central dataset with a representative sample of the world’s population. Reference values for BC parameters will be developed using the central dataset. The anticipated timeline for this project is 2-3 years. Our team and partners will work to make sure that the state-of-the-art study information generated is employed in developing awareness on the limitations BMI has as a diagnostic tool. The project will help spread the message that BF is more strongly associated with health outcomes compared to BMI while giving an insight into the public health importance of assessing the population’s BC. Finally, the study will help in structuring a universal BF classification that will be developed using a representative sample of the world’s population.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/body-composition-genetics-and-bone-outcomes

Body composition, genetics, and bone outcomes

Last updated:
ID:
52264
Start date:
15 January 2020
Project status:
Closed
Principal investigator:
Dr Victoria Bland
Lead institution:
University of Arizona, United States of America

Obesity and osteoporosis remain two major public health concerns. While obesity was once considered protective against osteoporosis, more recent evidence has shown an association between obesity and increased osteoporotic fracture risk. Inconsistency in findings may be due to the use of non-specific measures for obesity, such as body mass index (BMI) or percent body fat, which do not account for the physiological differences between adipose tissue depots. Fat tissue located around the organs, known as visceral adipose tissue, has been associated with metabolic dysregulation (e.g., chronic inflammation, insulin resistance) often seen in obesity. Meanwhile, fat tissue located directly under the skin in the abdomen and lower body regions, known as subcutaneous adipose tissue, is thought to contribute less to the metabolic dysregulation. Recently genetic studies have identified “favorable adiposity” gene variants that are associated with less visceral adipose tissue, greater subcutaneous adipose tissue, and better metabolic health.

The proposed study aims to address the gaps in knowledge regarding the association between soft tissue and bone health by analyzing the relationship between fat distribution, lean mass, and bone outcomes (e.g., bone mineral density, fracture history). This study will utilize data available from individuals in the UK Biobank imaging sub-study, including magnetic resonance imaging (MRI), dual energy x-ray absorptiometry (DXA), genetic, and health outcomes data. We also propose to use Mendelian Randomization, a genetic epidemiological technique, to further determine if there is a potential causal relationship between genes for body composition and bone outcomes. The proposed aims will advance our understanding of the relationship between body composition and bone health with the overarching goal to better predict individual risk for osteoporosis based on body composition and genetic profile.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/body-composition-lipodystrophy-and-metabolic-profiling-in-eating-disorders

Body Composition, Lipodystrophy and Metabolic Profiling in Eating Disorders

Last updated:
ID:
89892
Start date:
10 November 2022
Project status:
Current
Principal investigator:
Professor Nadia Micali
Lead institution:
Region Hovedstadens Psykiatri, Denmark

The aims of the present project are to analyse body composition and fat distribution in individuals with eating disorders from a large population-based study. We will also study the relationship between these parameters and grey and white matter in the brain. Lastly, we aim to evaluate the relationship between blood parameters related to metabolism, body composition, and brain structure in individuals from the UK BB with eating disorders and those without psychiatric or physical disorders.
Eating Disorders lead to severe medical complications, and researchers have shown differences in body tissues and distribution of fat tissues in those with eating disorders. For example, fat tissue distribution has been found to be more centrally-located, both in patients with anorexia nervosa and in patients with binge eating disorder, despite the fact that individuals with these disorders are at the two opposite extremes of body weight.
In a previous study we have shown that obesity is linked to abnormal brain network functioning, here we want to understand if fat tissue distribution might be differentially related to brain structural features, namely grey and white matter integrity and volumes in those with eating disorders and controls. Lastly, body tissue distribution is associated with laboratory parameters (for example, LDL and HDL cholesterol, fasting insulin, thyroid, sex, and stress hormones etc) in healthy and obese subjects, but this has not been studied in a large group of individuals with eating disorders from the general population. Findings from our study will improve our understanding of fat tissue distribution, physiology and metabolism in eating disorders and could allow better treatment of physical complications in this patient population.
The present project is expected to last roughly 12 to 18 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/body-fat-composition-and-risk-of-obesity-related-cancers-among-adults-with-normal-body-mass-index-in-the-uk-biobank-cohort

Body fat composition and risk of obesity-related cancers among adults with normal body mass index in the UK Biobank cohort

Last updated:
ID:
40525
Start date:
25 June 2018
Project status:
Closed
Principal investigator:
Dr Thomas Rohan
Lead institution:
Albert Einstein College of Medicine, United States of America

Over the years, it has been thought that being normal weight (i.e. having BMI between 18.5kg/m2 and 24.9kg/m2) is not associated with altered risk of obesity-related cancers such as breast, endometrial and colorectal cancers. Recently, this long-held view has been called into question, as this BMI category also includes individuals who possess abnormalities, such as excess body fat and high insulin levels, that can promote the development of cancer. Very few studies have, however, been conducted to explore whether risk of obesity-related cancers among normal size adults differs by levels of body fat. So far, the findings from existing studies suggest that, like overweight/obese adults, normal weight individuals with excess body fat may also have an increased risk of obesity-related cancers, specifically breast and colorectal cancer. Given the scarcity of studies, or lack thereof, in this area, we propose to undertake this study to improve our knowledge of the role of body fat composition in the development of obesity-related cancers among normal sized men and women. We will use statistical methods to assess the association between body fat and selected obesity-related cancers among the participants. As excess body fat also causes changes in levels of cancer-associated inflammatory/metabolic markers, we will also 1) assess the associations between the markers and body fat composition and 2) examine whether the association between body fat composition and the cancers is mediated by selected markers. We intend to complete this study within six months. This study can provide important information that will help to more accurately determine risk of obesity-related cancers among adults with normal BMI. Such information will also be valuable in developing strategies to reduce the risk of cancer development.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/body-measurements-prediction-from-retinal-funds-photographs-via-convolutional-neural-networks

Body measurements prediction from retinal funds photographs via convolutional neural networks.

Last updated:
ID:
51922
Start date:
30 August 2019
Project status:
Current
Principal investigator:
Dr Zongyuan Ge
Lead institution:
Monash University, Australia

1a: Retinal fundus photograph is an effective media to observe retinal abnormality. That is where ophthalmic organs and diseases can be visually detected. A recent study shows that the information like gender, age, blood pressure, body mass index could also be measured from the photograph via deep learning to some extent [1]. Obviously, this study demonstrates that there is rich information in the photograph and novel features of diseases could be discovered from the fundus image. The primary aspect of this project is to try to predict the vision-related measurements (such as visual acuity and spherical power) and health indicators (such as heart variability) from the fundus image via deep learning. This quantification of these measurements and indicators could further evaluate the risk of systemic disease.

1b: Our work aligns with UK Biobank’s stated purpose by improving the prevention and early detection of myopia, hypopsia and systemic disease.
1c: We will use the technology of machine learning and computer vision to train a model that could automatically predict the risks of myopia, arrhythmia and etc. by extract information from retinal fundus photographs.
1d:
We are requesting a complete dataset of left or right fundus images with paired measurements which are listed in our submission information.
Dataset size: 84,767 participants.

[1]. Poplin, R., Varadarajan, A.V., Blumer, K., Liu, Y., McConnell, M.V., Corrado, G.S., Peng, L. and Webster, D.R., 2018. Prediction of cardiovascular risk factors from retinal fundus photographs via deep learning. Nature Biomedical Engineering, 2(3), p.158.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/boosting-disease-detection-rate-by-characterizing-the-genetically-informed-normal-range-of-biomarkers-and-imaging-measures

Boosting disease detection rate by characterizing the genetically informed normal range of biomarkers and imaging measures.

Last updated:
ID:
42124
Start date:
21 November 2018
Project status:
Closed
Principal investigator:
Dr Nathan Scott White
Lead institution:
Multimodal Imaging Services Corporation, United States of America

We aim to study the feasibility of generating personalized norms for biomedical tests given the genetic information. Many biomedical assessments, such as memory test, blood counts, and imaging measures, have substantial variations attributable to individuals’ genetic background independent of disease process. However, current clinical practice is still accustomed to use one single reference norm for a given test, ignoring the potential background differences. To address this issue, we intend to study the applicability of personalized norms by using data from UK Biobank. We will examine how much information we can extract from the relationships between genotypes and values from biomedical assessments, including physical measures, cognitive functions, blood counts, urine assays, and imaging measures. We then determine whether the genetically corrected tests can help the early detection of illness. The disease status is determined through the registered record of health-related-outcomes in UK Biobank. We expect to conduct the project through 36 months to critically examine each of the biomedical tests available in UK Biobank. We envision that the results will facilitate the public health in the general population. The biomedical assessments can be further enriched by genetic information, enhancing the net benefit of biomedical tests.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/boosting-the-power-of-gwas-using-novel-statistical-tools

Boosting the power of GWAS using novel statistical tools

Last updated:
ID:
27412
Start date:
21 April 2017
Project status:
Current
Principal investigator:
Professor Ole Andreassen
Lead institution:
University of Oslo, Norway

This proposal seeks to apply UK Biobank data to study the genetic architecture of human traits using novel statistical tools. We aim to investigate the relationship between mental disorders and co-morbid diseases such as cardiovascular disease, cancer and metabolic disease (as well as protective phenotypes). Genome-wide association studies (GWAS) have successfully identified many genetic variants influencing complex human traits. However, the identified genetic variants only explain a small portion of the heritability of these traits. To improve discovery of genetic variants in complex human traits, we have developed statistical tools building on a Bayesian statistical framework. This proposal seeks to increase discovery of genetic loci influencing a range of human traits and disorders. Identifying genetic factors that confer risk or protect against health-related traits is critical for understanding the causal mechanisms underlying disease, and the causal relationship shared between clinical conditions. Improved gene discovery might inform the development of genetic prediction tools and ultimately improve treatment strategies for large patient groups. Hence, the proposed research is entirely congruent with the stated aim of UK Biobank ?to improve the prevention, diagnosis and treatment of illness and the promotion of health throughout society?. We will analyze the GWAS data on complex traits in the UK Biobank cohort using novel statistical methodology. Using software and computational tools we are able to enhance gene discovery by integrating GWAS data with additional knowledge about genetic variants, including their association in related traits or their genomic position. To assess the replicability (i.e. the robustness of the results) of the identified variants, we will evaluate their association in independent GWAS cohorts. Finally, the results may inform the development of novel genetic prediction tools. We would wish to study the full UK Biobank cohort.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/brain-age-versus-chronological-age-a-large-scale-reproducible-study

Brain age versus chronological age: a large-scale, reproducible study

Last updated:
ID:
59172
Start date:
27 May 2021
Project status:
Closed
Principal investigator:
Professor Alexander Selvikvag Lundervold
Lead institution:
Western Norway University of Applied Sciences, Norway

The human brain is constantly changing throughout the lifespan. A progression of brain substance loss is expected, as well as volumetric, morphometric and signal changes in a variety of structures. The biological aging of the brain therefore reflect the individual’s chronological aging.

So-called “brain age” models attempt to use this fact to predict an individual’s chronological age directly from imaging data. Earlier work has shown that machine learning methods can be used to construct accurate brain age models directly from neuroimaging data from healthy persons, e.g. MRI recordings of their brains.

In individuals with brain disorders, such as Alzheimer’s disease, one expects biological and cognitive deviations from normal aging. This can potentially be detected as a gap between the predicted age and the actual chronological age, the so-called “brain age gap”. This has also been shown to be relevant to other diseases and conditions, e.g. neuropsychiatric and neurodevelopmental disorders, obesity and traumatic brain injuries, and the potential clinical impact of being able to assess the risk of age-related disease has motivated a lot of research into the construction of brain age models.

In our project we will create a fast (i.e minutes) and accurate end-to-end pipeline for brain age prediction directly from MRI examinations, using state-of-the-art deep learning techniques.

Our investigations indicate that there are still significant challenges related to robustness of brain age models. In our preliminary studies we have constructed a brain age model trained on a large heterogeneous collection of data. By careful evaluation of the model performance in various experiments with different setups of training and test data, we have strong indications that enlarging the imaging data used for training the model would significantly improve its performance and robustness.

All the details necessary to reproduce and extend our findings will be made available to other researchers. The project will therefore provide a stepping-stone towards bringing brain age models closer to practical usefulness.

Our project is already well underway, and we aim to complete the main steps by Q2 2021.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/brain-ageing-model-identifying-neuroimaging-patterns-with-relevance-to-neurodegeneration

Brain ageing model: identifying neuroimaging patterns with relevance to neurodegeneration

Last updated:
ID:
100495
Start date:
30 April 2024
Project status:
Current
Principal investigator:
Dr Vesna Vuksanovic
Lead institution:
Swansea University, Great Britain

This project will model healthy brain ageing from routine brain scans. The study is motivated by the lack of understanding of why peoples’ brains age in different ways, and why some people are more likely to develop dementia as they get older, whilst others stay healthy or have only mild memory problems. The key component of our approach is in using statistical tools to examine healthy brain ageing based on patterns from routine brain scans. We will identify (i) those patterns shared between people as they age and (ii) map out those changes across the lifespan with potential relevance to dementia. By mapping out changes in the healthy brain, we will look for patterns that might predispose someone to a particular type of dementia. The advantage of our approach is that it will be developed on standard clinical brain images and therefore can be easily used in hospital or memory clinic settings. This project is planned for 3 years to gain the maximum amount of insight from the UK BioBank cohort.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/brain-aging-in-major-depressive-disorder-mdd-investigated-by-multimodal-magnetic-resonance-imaging-at-the-network-level

Brain aging in major depressive disorder (MDD) investigated by multimodal magnetic resonance imaging at the network level

Last updated:
ID:
99951
Start date:
18 January 2024
Project status:
Current
Principal investigator:
Dr Philipp Georg Saemann
Lead institution:
Max Planck Institute of Psychiatry, Germany

The aim of this project is to understand if depression which is a stress-related condition and highly prevalent in the population is associated with accelerated aging of the brain. First insights on this question have been gained from magnetic resonance imaging (MRI), but most analyses have not considered that depression is associated with other medical conditions and risk factors that influence aging. Another factor that influences brain aging through-out the life-span and needs consideration in such a study is early life adversity.
For this complex undertaking, the UK Biobank is an ideal platform as both psychometric and medical data along with MRI data are accessible. We will first study the association between age and brain anatomy and brain function in a large sample of healthy controls with no history of depression and no specific medical risk factors and disorders. As many data points altogether determine the aging status, we study this association by multivariate methods that consider hundreds to thousands of data values at the same time.
As a next step, we will use this model to predict the ‘brain age’ for a total of four clinical groups:
* First, patients with a lifetime history of a major depression (and likely associated medical risk factors and disorders).
* Second, patients with no depression but the same proportion of medical risk factors and disorders.
* Both these groups will be subdivided into to subgroups according to the degree of early life adversity.
As the brain is organized in networks and as there is increasing insight that pathological aging such as Alzheimer’s dementia affect specific networks more than others. One of the analysis principles of this study is therefore to study the brain’s age at the network level, more specifically, calculating not one ‘average’ brain age but each one per network and per MRI technique. Here, we will use three MRI techniques: One focusing on the brain’s macroscopic anatomy, one on its fiber connections (‘wiring’), and one focusing on its (resting) functional status.
Last, we also address if the functional state of the brain during a depressive state bears similarity with ‘advanced aging’, that is, if the brain looks ‘older’ during a depression than it actually is biographically. Naturally, repeated measurements of patients with depression might be useful to understand if these ‘advanced aging’ pattern is reversible, similar as clinically most affective and cognitive problems pale off after a depressive episode.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/brain-aging-polygenic-risk-score-prediction-of-neurological-phenotypes

Brain Aging Polygenic Risk Score Prediction of Neurological Phenotypes

Last updated:
ID:
48467
Start date:
7 May 2019
Project status:
Current
Principal investigator:
Dr Christin Glorioso
Lead institution:
NeuroAge Therapeutics, Inc., United States of America

Quality of life in old age is often compromised by dementia, mild cognitive impairment, increased depressive symptoms, and declining mobility. Having better predictive tools for identifying at risk individuals and also better therapies for slowing down brain aging would be greatly beneficial. The goal of our study over the next three years is to establish whether a genetic test we developed is useful in identifying people who are at risk for faster brain aging and neurological diseases. The test is a polygenic risk score (PRS) comprised of ~1000 brain aging risk single nucleotide polymorphisms (snps), which is the sum of small person-to-person variations in a select number of genes. The PRS was developed by identifying snps that in aggregate predict biological brain age in human postmortem brain cohorts. We determined the biological age of the postmortem brains using RNA levels of ~1000 genes that change with normal aging throughout lifespan. Some subjects had biological brain ages very close to their chronological ages and some had brain ages that were older or younger than their chronological ages. We were able to correlate the difference between biological and chronological age (delta age) with genetic variation across the genome in meta-analysis. The aggregate number of risk snps for an individual is the basis for the PRS. In the UK Biobank, we will test whether this score can predict cognitive decline and neurological disease in large living cohorts. This will shed light on whether this test will be useful for physicians to put into place screening and preventative measures for at risk individuals. This may also help identify avenues for new drugs that will slow down brain aging, treat neurological diseases such as Alzheimer’s, as well as identify people who would most benefit from these treatments before they have become irreversibly impaired.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/brain-allometry-and-asymmetries-as-intermediate-phenotypes-between-genetic-and-environmental-factors-and-cognitive-function-and-mental-health

Brain allometry and asymmetries as intermediate phenotypes between genetic and environmental factors, and cognitive function and mental health

Last updated:
ID:
46007
Start date:
30 May 2019
Project status:
Closed
Principal investigator:
Professor Franck Ramus
Lead institution:
L'Ecole Normale Superieure, France

Humans are incredibly diverse at cognitive and behavioural levels. Understanding the factors that make each of us a unique human being is of major interest, both for fundamental science and for application to cognitive and mental disorders. Ultimate factors lie within the genome and the environment. While it is possible to find reliable associations between such factors and cognitive function, the relationships are long and indirect. Much understanding could be gained by considering intermediate factors between genes and cognition. The obvious candidates are within the brain, which collects both genetic and environmental influences, and is the biological basis for cognition, behaviour and mental health.
Although all humans share a common brain structure and organisation, they also vary enormously in terms of the size and shape of their brain and of its subcomponents. We will therefore focus on describing and understanding this variation in brain anatomy and its relationship with other factors. We will examine the genetic and environmental factors that give our brain a unique anatomical configuration. Furthermore, we will inquire, to what extent do certain brain anatomical features explain variations in cognitive functions, and constitute a risk for certain cognitive or mental disorders?
The proposed research will therefore contribute to illuminating the network of factors and the complex causal pathways that lead to cognitive deficits and mental disorders such as depression, anxiety, or schizophrenia.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/brain-anatomy-and-substance-use

Brain anatomy and substance use

Last updated:
ID:
40830
Start date:
31 August 2018
Project status:
Current
Principal investigator:
Professor Todd A Hare
Lead institution:
University of Zurich, Switzerland

We aim to break new grounds in the understanding of the relationship between substance use and brain anatomy. Our study will combine the available brain images in the UKB with information about substance use and genome-wide data.
The insights from our research will advance the understanding of the biological mechanisms underlying substance use, and may identify targets of novel treatments and interventions. Importantly, our research design will combine neuroanatomical measures with behavioral and genetic data that will allow addressing questions regarding the direction of causal relationships between brain anatomy and substance use. Furthermore, our findings may yield powerful biomarkers that could be useful for a number of purposes including the identification of individuals at risk before the onset of pathological behavior, providing possibilities for more targeted therapy and prevention strategies. The biomarkers our study will identify can also be used as control variables in randomized control trails, reducing the costs of such trails by decreasing the number of required participants to achieve a given level of statistical power. Finally, the results of our study will contribute to precision medicine and provide new tools to study how specific individuals are expected to react to policies aiming to reduce substance abuse.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/brain-asymmetry-and-the-corpus-callosum

Brain asymmetry and the corpus callosum.

Last updated:
ID:
199592
Start date:
8 October 2024
Project status:
Current
Principal investigator:
Miss Caitlin Dale
Lead institution:
University of Auckland, New Zealand

The two sides of the brain in popular psychology are often used to stand in for particular cognitive modes, with the ‘left-brain’ responsible for everything analytical, rational and linguistic and the ‘right-brain’ responsible for creativity and emotion. Although this is certainly an oversimplification, the brain does possess regions specialised for different functions, such as language processing and facial recognition, and these are undoubtedly distributed differently over its two sides. But how might these functions be reflected in the structure of the brain? Or, put conversely, how might brain structure enable these broader cognitive modes? Even with respect to language processing, one of the most typically ‘left-brained’ functions, this relationship remains unclear.
Moreover, our brains are massively interconnected. The corpus callosum is our major cerebral information highway, linking the two sides of the brain with more than 200 million nerve fibers. How might the asymmetry of the ‘left’ and ‘right’ brains relate to the means of communication between them? It is conceivable that as humans evolved, we needed to use our limited brain size efficiently by developing more and more specialised brain functions. We might expect that less communication was consequently needed between the sides of the brain, because local connections were favoured as more economical. In an alternative view, integrating across these widely spread-out specialised regions might require an information highway with even more ‘lanes’. These are two possibilities, but more research is required to illuminate the nature of the relationship between brain asymmetry and the brain’s major communication pathway.
It is important to establish what this relationship might look like in the general population, as brain asymmetry has been implicated in diverse conditions such as autism, obsessive compulsive disorder, schizophrenia and Alzheimer’s disease. Such a baseline could help researchers identify where the brain differences lie in these conditions, enhancing clinical understanding and informing possible treatment approaches.
This project will run for three years (36 months) and will seek to outline any links between brain asymmetry and the corpus callosum, how these might relate to broader cognitive functions and what factors might play a role in how they are expressed.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/brain-based-biomarkers-of-accelerated-aging-across-the-lifespan

Brain-based biomarkers of accelerated aging across the lifespan

Last updated:
ID:
493261
Start date:
14 January 2025
Project status:
Current
Principal investigator:
Professor Ahmad Hariri
Lead institution:
Duke University, United States of America

The proliferation of brain-based aging biomarkers has generated optimism that MRI could be useful in gauging individual risk for aging-related functional decline and chronic diseases. However, it is unclear how existing brain-based biomarkers compare to one another in predicting decline and disease. To be nominated for clinical adoption within the precision medicine model, we need to better understand how different brain-based biomarkers map onto different types of chronic aging-related disease risk and how they relate to known risk factors for aging-related disease and decline.

Ideally, MRI-derived brain-based biomarkers of accelerated aging are first identified in one dataset to predict a health-related outcome and then generated in an independent dataset to establish predictive utility. Here, brain-based biomarkers of accelerated aging identified in other studies will be derived in the UK Biobank dataset to test each biomarkers’ sensitivity to disease and decline. We will also compare brain-based biomarkers with non-brain-based biomarkers of accelerated aging including various -omic clocks. Furthermore, we will map the differential associations of these biomarkers onto established risk factors for aging-related decline and disease including genetics, experience, environment, socioeconomics, lifestyle, and health.

Identifying which specific brain-based biomarkers or combination of biomarkers is most effective at predicting specific outcomes will help guide the prioritization of specific biomarkers for clinical translation.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/brain-basis-of-visual-imagery-revealed-by-big-data

Brain basis of visual imagery revealed by big data

Last updated:
ID:
96049
Start date:
6 November 2024
Project status:
Current
Principal investigator:
Dr Timo L Kvamme
Lead institution:
Aarhus University, Denmark

We use mental imagery in many activities in everyday life, such as when we are going from A to B, we use our mental images to imagine our route. We can also imagine how our work will end and thus plan according to that image. The ability to have mental images and to report on these is also what makes us uniquely human. Researchers have begun to investigate these so-called “higher-order” cognitive functions like mental imagery. One research question is currently how mental image relates to similar cognitive functions like remembering a person’s face or our sense of direction. Another research question considers the biological underpinnings of the brain – the hardware underlying these cognitive functions. Our project aims to enrich our understanding of the human capacity for mental imagery.

The project aims to understand the neurobiological underpinnings of certain individuals’ inability to create mental imagery and of the opposite, those who fail to distinguish imagery from perception. These traits may relate to clinical disorders such as schizophrenia, which are devastating for the families of those affected and create an incredible burden on national economies. The understanding provided through our research will help point to further clinical research projects.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/brain-changes-associated-with-pregnancy

Brain changes associated with pregnancy.

Last updated:
ID:
55993
Start date:
23 March 2020
Project status:
Closed
Principal investigator:
Dr Susanna Carmona
Lead institution:
Fundacion para la investigación Biomedica del HGM, Spain

Pregnancy involves radical hormone surges and biological adaptations that can lead to health problems. However, the effects of pregnancy on the human brain, and its implication to psychopathology remain understudied. In a previous study, we showed that pregnancy renders substantial gray matter reductions in regions involved in maternal behavior.
The project proposed in this application aims to gain insights into the way a woman’s brain (as assessed by different MRI techniques) changes during pregnancy. In particular, we want to test whether brain changes are restricted to the first years of motherhood or whether they endure for several years. We also aim to test if brain changes associated with pregnancy are related to gestational factors (number of previous pregnancies, and number of babies born from each pregnancy), and to an increased risk for the development of mental disorders.

We will compare different brain metrics (anatomy, structural connectivity, and functional connectivity) across three main groups: nulliparous women, primiparous women, and multiparous women. We hypothesize that: 1) mother (primiparous and multiparous) will have less gray matter volume in regions involved in maternal behaviors, and 2) mothers will differ from nulliparous women in metrics of functional and structural connectivity. In addition, we want to explore whether there is an association between brain differences (mothers vs. non-mothers), and gestational, cognition, and psychiatric factors.

This project represents a challenge for advancing in the scientific understanding of one of the most important dimensions of a living being: motherhood. Its results can impact different clinical areas, such as neurodevelopmental and neurodegenerative conditions, reproductive biology, psychiatry, and developmental psychology.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/brain-disease-models-using-multivariate-causality

Brain disease models using multivariate causality.

Last updated:
ID:
84616
Start date:
21 March 2023
Project status:
Current
Principal investigator:
Dr Ferath Kherif
Lead institution:
Centre Hospitalier Universitaire Vaudois, Switzerland

People are not all equal when it comes to conditions such as mental illness, since there are a number of factors which are directly or indirectly related to the symptoms and disease progression. Therefore, it is crucial that we create a model which takes all of those factors into account. Now this is possible due to the availability of large scale data and the power of multivariate approaches for explaining individual differences. During this study, we hypothesized that these data could be summarized into a few components that are age and gender dependent, based on the few components, we could develop causal brain health models.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/brain-disease-subtyping-modeling-aging-and-subtyping-brain-disease-in-multimodal-brain-mri

Brain Disease Subtyping Modeling aging and subtyping brain disease in multimodal brain MRI

Last updated:
ID:
46608
Start date:
12 March 2019
Project status:
Closed
Principal investigator:
Dr Owen Robert Phillips
Lead institution:
Brain Key Inc, United States of America

Overall, this research project aims to better understand how the brain ages. This is important because if we can better understand the normal brain aging process, then we can more quickly identify when patients with brain disorders deviate from this process. The hope is if we can identify a patient early in their disorder, then perhaps we can better treat the disease before it progresses. Patients with both neurodegenerative diseases such as Alzheimer’s disease and psychiatric disorders such as schizophrenia have both been shown to have abnormal brain aging so a greater understanding of the processes involved in this abnormal aging may help in both early detection and the identification of novel biomarkers.

In order to develop a model of how the brain ages, this project it will focus on the multimodal brain imaging data that was collected as part of the UK Biobank project. The neuroimaging models will be refined by integration with cognitive and genomic data.

Additionally, this research seeks to develop models for subtyping brain disease. Subtypes of brain disease are often hard to identify early, which can result in additional testing and delay optimized treatment. Finally, this project will also investigate potential links between cardiac and brain health. Up until now, there has been limited research into the connection between cardiac and brain health because of limited data and computational resources to pursue a link between the two. This aim is highly exploratory but if reliable links between cardiac and brain health can be established, it may help in developing effective treatments. This project is expected to take two years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/brain-health-research-using-bhq-an-international-standard-index

“Brain health research using BHQ, an international standard index”

Last updated:
ID:
103988
Start date:
18 October 2023
Project status:
Current
Principal investigator:
Dr Yoshinori Yamakawa
Lead institution:
Kyoto University, Japan

This project aims to find out how social, psychological, and physical factors affect the health of our brains. We will do this by analyzing open MRI and questionnaire datasets. The research period will be from April 2023 to March 2025. We believe that our findings will help people better take care of their brains and potentially help prevent or slow down dementia.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/brain-injury-after-covid-19-infection-biomarker-study-biacob

Brain Injury After COVID-19 infection Biomarker Study (BIACOB)

Last updated:
ID:
76059
Start date:
16 September 2022
Project status:
Current
Principal investigator:
Professor Paul M Matthews
Lead institution:
Imperial College London, Great Britain

Our project will use very small amounts of blood from UK Biobank participants in the COVID re-imaging studies to assess markers of brain injury in those exposed to SARS-CoV-2 relative to those who have not had this exposure. This biomarker data would be linked to other clinical, imaging, lifestyle and exposure data to explore factors that could be responsible for variations in responses of different people.

Severe infections may cause dementia, but this has been difficult to test. The observation that brain symptoms with COVID-19 are common, particularly in people with clinically severe presentations, suggests that studying the brains of those exposed to SARS-CoV-2 could test this. Additional data available in UK Biobank also would allow exploration of potential contributions to any brain injury that could be due to SARS-CoV-2 effects on other organs, such as the heart or blood vessels. Finally, as the effects of SARS-CoV-2 exposure may depend on the genes that a person inherits, we will relate results of these analyses to individuals’ genetic makeup.

Assays for the blood markers will be completed within 6-7 months of the end of the UK Biobank COVID re-imaging (UKBB-CRE) study. We will make our biomarker results available to all researchers. We are experienced in analyses of these data and will be conducting our analyses quickly to accelerate the open data release date.

Meeting our goals will allow us to test for clinical unsuspected, “preclinical” brain injury from SARS-CoV-2 and to define factors related to it. The specific plasma biomarkers chosen will allow testing whether COVID accelerates late life dementia/Alzheimer’s disease, whether brain pathology related to COVID can explain persistent brain symptoms (e.g., “brain fog”) and how brain and body changes after infection might explain other symptoms of Long COVID. Together, this information will have public health impact for defining populations at risk of dementia after SARS-CoV-2 infection and suggest therapeutic approaches.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/brain-iron-and-systemic-inflammation-in-neurodegenerative-disease-brind-study

Brain iron and systemic inflammation in neurodegenerative disease (BRIND) study

Last updated:
ID:
189701
Start date:
6 December 2024
Project status:
Current
Principal investigator:
Professor Ian Galea
Lead institution:
University of Southampton, Great Britain

Multiple sclerosis (MS) and Alzheimer’s Disease (AD) are common neurodegenerative disorders in younger and older adults respectively. While they have different causes, they are both associated with damage to the brain. Also certain factors, such as systemic infections may make both conditions worse. An increased amount of iron in the brain has also been linked with both MS and AD. Recent research has found that mild systemic infections may result in the release of haemoglobin, the red pigment in red blood cells, and this is a source of iron. Hence it is possible that there is a link between systemic infection, red blood cell release of haemoglobin, brain iron and severity of the condition.

Several thousand UK Biobank participants have a diagnosis of MS or AD. Control individuals with no neurological disease are easily selected from amongst the other participants. In this project the brain iron content of individuals with AD and MS will be studied to see whether there is a link to haemoglobin release and/or systemic inflammation, and disease severity. The severity of MS or AD will be based on patient report as well as brain magnetic resonance images. Patient report will include memory and information processing speed, physical activity, employment, and other social and health outcomes.

This study may deliver new insights which may in turn lead to new treatments for MS or AD. It will provide a PhD student, working under close supervision, with experience in research and scientific publications to further their careers.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/brain-mechanisms-underlying-symptom-perception-in-asthma

Brain mechanisms underlying symptom perception in asthma

Last updated:
ID:
25673
Start date:
1 February 2017
Project status:
Closed
Principal investigator:
Dr Kyle Pattinson
Lead institution:
University of Oxford, Great Britain

This research aims to understand the brain networks underlying symptom perception in asthma. In up to 60% of people with asthma, symptoms may be `inaccurate` or discordant – meaning that symptoms poorly (but variably) reflect medical markers of lung function and inflammation. Discordant symptoms are associated with emotional stress and with worse asthma outcomes, leading to increased suffering and use of healthcare resources.

Our recent work suggests that brain networks associated with emotion communicate with those that regulate breathing and breathlessness. With the Biobank brain imaging data, we would test whether dysfunction in these pathways explains discordant asthma symptoms. Asthma affects 5-10% of the UK population, and is associated with £1.2 billion health care costs p.a. in the UK. The proposed research will help improve the diagnosis and treatment of asthma in the following ways:

1) Better phenotyping: Asthmatics with the most discordant symptoms represent a `difficult to treat` and poorly understood group, and the Biobank brain imaging data gives an unparalleled opportunity to understand which brain networks may be aberrant in these people.

2) New targets for individualised treatment based upon brain biomarkers – e.g. new and repurposed therapies that may target networks identified above. Brain scans will be analysed and compared to both clinical measures (lung function and blood counts) and psychological measures (anxiety, depression, and breathlessness).

The asthmatic participants will be compared to a control group of participants, and will also be stratified to investigate whether groups of asthmatics can be identified whose breathlessness is more susceptible to psychological factors or to clinical disease measures.

Their brain structure and function will be analysed to see whether these participants have noticeable differences in how areas of the brain ?communicate? with each other, and in particular those areas involved with emotional regulation and symptom perception. The participants to be included in this research would be all participants (i.e. ~11,000) in the Biobank that have undergone brain imaging.

From this we will select cases of asthma (~1,200 participants) and carefully matched controls without asthma (matched for variables including age, sex, socioeconomic status, education, comorbidities, drug therapy etc).


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/brain-morphometry-and-functional-network-connectivity-across-sexes-in-the-healthy-ageing-population

Brain morphometry and functional network connectivity across sexes in the healthy ageing population.

Last updated:
ID:
107123
Start date:
7 March 2024
Project status:
Current
Principal investigator:
Professor John P John
Lead institution:
National Institute of Mental Health and Neuro Sciences, India

We aim to investigate if sex plays a significant role in structure and functional connectivity of the brain in a healthy aging population of Indian origin.Using structural and functional magnetic resonance imaging (MRI), we will measure brain size, volume, and form, as well as the strength of association between and within brain regions with regard to functionality. Previous research conducted in our lab emphasizes the importance of considering sex as a contributing factor in brain data analysis.We would like to explore participant data of Indian origin as there is limited research in this area. Other influencing factors such as age, total brain volume, education, biochemistry (role of hormones), handedness and physical activity would be used as nuisance variables. We’ll use a machine learning algorithm to train the dataset at our lab and test the UK Biobank data using the features obtained while training (The algorithm learns from known data to make predictions or classifications on new unseen dataset. )
Understanding potential sex related differences with or between the left and right brain functions can shed light on various cognitive abilities such as memory, attention, creativity etc. It can also reveal any influences of structure on functional connectivity between sexes.Our previous study on left and right brain dominance between sexes, we found that the idea that men are more logical and analytical which is associated with the left brain function while women are more intuitive and creative which is associated with the right brain functions. This breaks stereotypes and identifies the unique capabilities of each sex. Stronger connectivity in specific brain regions in males or females may be associated with cognitive or behavioral aspects more pronounced in that respective sex.
The influence of biological sex brings numerous benefits to communal health. It allows for the identification of sex-specific health risks, evidence-based health policies, and equal access to healthcare. Recognizing the impact of biological sex on disease prevalence and outcomes enhances disease surveillance efforts, leading to more effective interventions and prevention strategies. Furthermore, the unique needs and strengths associated with each sex allows for informed parenting practices and educational approaches, promoting optimal child development and reducing gaps. Incorporating this knowledge into social program strategies helps address sex-specific issues and better support individuals. Exploring brain differences between sexes can also guide interventions for mental well-being and detect modifiable changes if any.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/brain-shape-analysis-from-very-large-datasets

Brain Shape Analysis from very Large Datasets

Last updated:
ID:
20576
Start date:
1 June 2016
Project status:
Current
Principal investigator:
Professor Nicholas Ayache
Lead institution:
Inria, France

An extensive study of the brain anatomy could lead to powerful biomarkers to detect brain diseases affecting the brain shape. The UK Biobank initiative offers a unique opportunity to build for the first time a truly extensive study of the brain over a significantly large cohort. Our proposal is threefold: (1) to identify possible methodological bottlenecks in processing very large datasets of brain images, (2) to provide a new state-of-the-art statistical atlas across ages and populations, and (3) to further study possible links between brain anatomy and biological criteria, such as age, gender, and other factors. The study of the brain anatomy, with its variation across ages and populations, is key for establishing a precise characterization of normal evolutions in healthy subjects. Any deviation from such established standard is a possible risk indicator, for instance, of the Alzheimer?s disease. Research on building and exploiting statistical atlases from very large datasets potentially improves diagnostics and follow-ups in brain diseases affecting the brain shape. A statistical atlas will be built using the neuro images from the UK Biobank participants. The average shape of the brain, and its variations, will be studied by considering possible links with several biological criteria, such as age, gender, and other factors. Full cohort.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/brain-structural-markers-and-clinical-features-of-understated-cognitive-impairment-uci-in-middle-age-45-65-a-cohort-study

Brain structural markers and clinical features of Understated Cognitive Impairment (UCI) in middle age (45 – 65): a cohort study

Last updated:
ID:
147797
Start date:
21 March 2025
Project status:
Current
Principal investigator:
Mrs Marine Lunven
Lead institution:
Université Paris Est Créteil, France

Understated Cognitive Impairment (UCI) corresponds to a significant deficit in cognitive performance in the absence of subjective cognitive complaints (SCCs). A significant proportion (30%) of 50-65 years old had pathological cognitive performance in the absence of SCCs associated with cardiovascular risk factors, obesity, and metabolic syndrome. These factors and UCI has already been reported to be related to a higher risk of cognitive decline and dementia. However, no study of UCI in this age group has yet used a comprehensive battery of cognitive tests to determine whether domains other than the executive one are impacted. Advanced automated MRI methods have emerged as more objective and precise alternative to assess brain damage related to neurological deficits.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/brain-structure-and-connectivity-links-to-metabolic-health-and-diabetes

Brain structure and connectivity – links to metabolic health and diabetes

Last updated:
ID:
104017
Start date:
19 October 2023
Project status:
Current
Principal investigator:
Professor Martin Heni
Lead institution:
Ulm University, Germany

Our body’s metabolism is regulated by a complex interaction between the brain and peripheral organs, such as the liver, muscles, and adipose tissue. Research over the years has shown that the human brain can regulate metabolism in the peripheral organs through specific signaling pathways. Impairments in these regulatory mechanisms can contribute to the development of metabolic diseases such as diabetes and obesity.

In a new analysis of UKBB data, we aim to investigate the links between the core hubs of brain anatomy, structure, and connectivity focused on the hypothalamus which are linked to metabolism, focusing on pre-diabetes and diabetes, including novel subgroups of diabetes. We will also investigate adiposity and body fat distribution in a cross-sectional as well as a longitudinal manner. Our research will involve linking brain imaging data from the UK Biobank, to clinical, laboratory, and genetic data. We will explore the functional and anatomical connections between brain regions involved in metabolic regulation, with a particular focus on the hypothalamus, which plays a critical role in regulating glucose and energy metabolism.

The research will also investigate how environmental factors such as diet and exercise modulate links between the brain and whole-body metabolism. This information could be valuable in developing interventions aimed at promoting healthy lifestyles and preventing metabolic diseases.

Our analyses are of significant public interest as metabolic diseases are a growing public health concern worldwide, affecting millions of people and placing a significant burden on healthcare systems. The research has the potential to advance our understanding of metabolic diseases and could contribute to new approaches for their prevention and management. It could ultimately contribute to improving public health and reducing the burden of metabolic diseases on individuals and healthcare systems.

The UK Biobank is a valuable resource for this research, providing a large and diverse cohort of participants, including individuals with varying degrees of metabolic health. By linking brain and whole body magnetic resonance imaging data to clinical, laboratory, and genetic data from this cohort, our research has the potential to provide a more comprehensive understanding of the complex interplay between brain metabolism and peripheral organs.

In summary, our research aims to provide critical insights into brain mechanisms underlying metabolic diseases such as diabetes and obesity.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/brain-structure-and-function-in-mental-health-a-focus-on-self-harm-suicide-dementia-risk-and-trauma-related-disorders

Brain Structure and Function in Mental Health: A Focus on Self-harm, Suicide, Dementia Risk, and Trauma-Related Disorders

Last updated:
ID:
733840
Start date:
26 June 2025
Project status:
Current
Principal investigator:
Dr Sun Jae Jung
Lead institution:
Yonsei University, Korea (South)

Our research aims to investigate the relationships between brain structure, function, and mental health disorders through studies utilizing UK Biobank’s extensive neuroimaging data.
First, we will examine the neurobiological basis of self-harm and suicidal behaviors. While previous research has explored brain-behavior relationships in suicidality, findings have been inconsistent due to limited sample sizes. To establish a clearer understanding of these associations, we will identify genetic factors influencing brain structure and function and examine their potential causal relationship with self-harm and suicidality. Additionally, we will track longitudinal outcomes following self-harm incidents to understand how different self-harm methods and healthcare interventions influence patient outcomes, advancing our understanding of these behaviors and informing intervention strategies.
Second, we will investigate the neural mechanisms underlying the increased dementia risk observed in individuals with mental disorders. Current evidence suggests that brain structural and functional alterations may mediate this relationship. However, these findings have been largely derived from cross-sectional studies, limiting causal inference. Our longitudinal analysis will examine how mental disorders contribute to dementia risk through specific patterns of brain changes, potentially identifying early markers and intervention targets.
Third, we will explore sex-specific associations between sexual trauma and amygdala structure and function. Previous studies have suggested that traumatic events, such as sexual violence, may lead to structural and functional abnormalities in the amygdala, which may contribute to the development of psychiatric disorders. However, findings are inconsistent due to limited power and consideration of sex differences. Our analysis will leverage UK Biobank’s large sample size to conduct sex-stratified analyses of trauma-related amygdala changes.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/brain-substrates-linking-gastrointestinal-symptoms-and-psychiatric-disorders

Brain substrates linking gastrointestinal symptoms and psychiatric disorders

Last updated:
ID:
49555
Start date:
9 March 2020
Project status:
Closed
Principal investigator:
Professor Joseph LeDoux
Lead institution:
New York University, United States of America

Anxiety and depression are significantly increased in people with gastrointestinal disorders. Although gastrointestinal symptoms have been long linked to altered psychological factors, it is poorly understood today if and how these disorders intertwine at the brain level. One limitation involved when studying these disorders is that there is a heterogeneous manifestation of symptoms across the relevant population. In this study, we sought to look for brain correlates of specific gastrointestinal symptoms. Once differential brain patterns are detected, we will look for associated anxiety and depressive symptomatology. The aim is to find distinct clusters of mixed psychological and gastrointestinal symptoms that are prevalent in our population. We will then behaviorally characterize mixed-symptomatology clusters in relationship with cognitive functions and emotional processing. This work will allow us to detect biologically relevant clinical pictures and facilitate future investigations. This study will also advance our mechanistic understanding of complex psychiatric pathologies and has the potential to inform the development of new treatments.

Lastly, we will look for environmental factors that may foster or hinder symptom-clusters. Factors like diet, sleep, and early-life stress have been related to the occurrence of anxiety and depression, as well as to the occurrence of gastrointestinal disorders.

We believe that our work, both the analyses and the publication, will be complete within two years


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/brain-volume-atlas

Brain volume atlas

Last updated:
ID:
25694
Start date:
2 January 2017
Project status:
Closed
Principal investigator:
Keith Goatman
Lead institution:
Toshiba Medical Visualization Systems Europe Ltd, Great Britain

The research will investigate computer software for measuring abnormal changes in particular brain region volumes, seen in magnetic resonance images. The specific regions to be investigated are associated with diseases such as dementia, for example Alzheimer’s disease. A database of normal brain region volumes will be built, using automated software we have developed, based on a reference brain region atlas developed at Johns Hopkins University. The research would likely form part of a clinical/research software application to help diagnose degenerative brain disease, for example Alzheimer’s Disease. It could also be used to quantitatively assess the physical effects of treatments for degenerative diseases. In order to detect abnormal brain regions we will first create a database of normal brain volumes. It will be based on equal numbers of male and female subjects from the Biobank imaging cohort, evenly spread across the participants’ age range. Automatic software will be used to measure the volumes of specific brain structures known to be affected by degenerative diseases. We will validate the performance using subjects with normal memory scores, subjects with abnormal memory scores, and subjects known to have dementia from another study. 1000 subjects will be chosen from the imaging cohort, selected by stratified sampling of the three age decades, with equal male/female ratios throughout age range. 800 subjects would be selected with normal memory test scores, and 200 with abnormal memory scores. Note that subjects may be excluded based on a previous history of neurological disease or symptoms other than the memory test.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/brain-white-matter-anomalies

Brain white matter anomalies

Last updated:
ID:
18359
Start date:
6 April 2018
Project status:
Closed
Principal investigator:
Dr Fabrice Crivello
Lead institution:
University of Bordeaux, France

Detect and quantify on brain MRI different types of white matter anomalies (hyper signals, perivascular spaces). Assess effects of demographic, clinical and environmental variables on the number and volume of these anomalies. White matter anomalies are thought to be biomarkers of small vessel diseases and cognitive decline. The proposed research will provide key elements on the lifespan course and risk factors of white matter anomalies appearance and development which will provide arguments for developing new prevention/intervention strategies against age-related brain pathologies. We will develop an automated computer analysis of brain MRI scans searching for anomalies in the white matter tissue and study statistical association of the quantity and volume of these anomalies with demographic, lifestyle, and bioclinical variables 10,000 (all available UK biobank participants with brain MRI data)


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/bridge-biologically-structured-representations-for-interpretable-deep-genotype-phenotype-exploration

BRIDGE: Biologically-structured Representations for Interpretable Deep Genotype-Phenotype Exploration

Last updated:
ID:
880359
Start date:
5 August 2025
Project status:
Current
Principal investigator:
Mr Chrysanthos Christou
Lead institution:
Aristotle University of Thessaloniki, Greece

We will build BRIDGE, a new computer tool that learns how our genes influence simple traits-like eye or hair colour-and complex conditions such as schizophrenia. BRIDGE will take genetic information from UK Biobank volunteers and look for patterns that predict these traits.

To make sure our tool is both accurate and easy to understand, we will compare two ways of building it:

A “guided” version that follows known gene relationships and looks for small blocks of related variants.

A “brute-force” version that starts by examining every possible connection and then trims away the least important ones.

By testing both approaches side-by-side against an existing tool called GenNet, we will see which method gives the best predictions and the clearest insights into which genes and gene groups matter most.

Our work will:

Help researchers and doctors see exactly which genes drive each trait, rather than treating the computer model as a “black box.”

Offer new clues about how many different genetic factors combine to influence health and appearance.

Share our BRIDGE code and easy-to-read summaries with the wider community, so others can build on our findings.

This project uses UK Biobank data to advance transparent, trustworthy genetic research that benefits everyone.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/bridging-research-with-multi-scale-biomarkers-and-neuropsychiatric-symptoms-based-on-theory-and-data-driven-approaches

Bridging research with multi-scale biomarkers and neuropsychiatric symptoms based on theory- and data-driven approaches

Last updated:
ID:
90808
Start date:
21 July 2022
Project status:
Closed
Principal investigator:
Dr Yuta Takahashi
Lead institution:
National Center of Neurology and Psychiatry, Japan

Neuropsychiatric disorders are a major burden for many people. Many studies have been conducted to investigate relationships between various biomarkers, such as genomics and brain MRI, and neuropsychiatric disorders, and to explore pathogenic mechanisms of neuropsychiatric disorders. However, relationships between markers and symptoms are highly complex and are still unknown. It has also been suggested that treatment responsiveness varies even within the same disease, and that there are common pathogenic mechanisms shared by multiple neuropsychiatric disorders, suggesting the need for a new disease classification. Our goal is to explain the complex relationship between biomarkers and symptoms by combining two artificial intelligence technologies for various neuropsychiatric disorders, and to determine the complex pathogenic processes of these diseases, which have not been revealed by conventional analysis. One of the two artificial intelligence technologies is a theory-driven approach that prepares an artificial neural network analogous to information processing of the human brain to reveal changes in information processing that occur in neuropsychiatric diseases. The other artificial intelligence technology is a data-driven approach that uses machine learning on big data to reveal hidden associations among the genome, brain MRI, and neuropsychiatric symptoms that cannot be found using conventional methods. Combining these technologies, we have prepared an artificial neural network for each subject and simulate developmental process leading to the onset of neuropsychiatric disorders in that subject. This research is expected to clarify developmental processes that cause neuropsychiatric disorders and to understand neuropsychiatric disorders from the perspective of information processing in the brain. This will enable application to therapies that regulate neural circuits in the brain, and is expected to contribute to creation of new disease classifications and personalized medicine. The research period is planned to be 36 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/building-a-global-health-foundational-model-using-generative-ai-algorithms

Building a Global Health Foundational Model using generative AI algorithms.

Last updated:
ID:
240581
Start date:
8 May 2024
Project status:
Current
Principal investigator:
Dr Thomas Stubbs
Lead institution:
Chronomics Ltd., Great Britain

Generative Artificial Intelligence (AI) algorithms have already revolutionised our lives across domains such as text, image and video. For example, ChatGPT is one of the most successful technology launches of all time, helping millions of users around the world to increase productivity and brainstorm ideas through text generation. Applications like ChatGPT are built on top of Foundational Models, which have been trained in huge datasets and can be used as building blocks or pillars for downstream applications.

Our healthcare systems are currently mired in complexity, often relying on disjointed data, low interoperability, high administrative burden and slow innovation. The potential of AI to disrupt healthcare is tremendous, but there is a need to create a Foundational Model that is suitable for healthcare applications.

In this project we will develop the world’s first Global Health Foundation Model by leveraging the latest methods in AI algorithms, huge computational power and infrastructure and combining de-identified health data from millions of volunteers across diverse datasets (including the UK Biobank). With this model, we aim to synthesize and understand health data on an unprecedented scale and to be able to generate synthetic health data with high fidelity while ensuring the highest standards of privacy and security.

All of this will lead to a transformation in healthcare delivery, risk assessment, and personal health insights. The applications of the Global Health Foundation Model are probably beyond what we can imagine, such as helping patients to better understand their medical results (e.g. imagine that you could talk with your Electronic Health Record in plain language) or supporting doctors to make better treatment decisions (e.g. by contextualising the latest laboratory test or radiology image, producing more accurate diagnoses and predicting the future evolution of a patient).

This research will benefit the health and wellbeing of society. By reducing the time and costs associated with health data access, we will enable more people to access and analyse data-driven insights (inc. researchers, innovators, medical professionals or epidemiologists). Furthermore, by providing the foundational block to train other models for specific healthcare applications, we will ultimately accelerate the impact of health advancements. Finally, by training the model on data from individuals with different genetic backgrounds and environmental contexts, we will ensure health equity is achieved and no one is left behind.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/building-a-multi-disease-prognostic-panel-toward-individualized-preventive-medicine

Building a multi-disease prognostic panel toward individualized preventive medicine

Last updated:
ID:
105777
Start date:
9 August 2023
Project status:
Current
Principal investigator:
Mr Jacob Vogel
Lead institution:
Lund University, Sweden

Many age-related disease progress silently in the body without generating any symptoms. Once symptoms do appear, the disease are often in a more advanced stage and is harder to treat. In theory, being able to recognize, diagnose and track diseases early on would give health professionals a better chance at successfully treating diseases in a cost effective way. Unfortunately, we do not have good ways of tracking and diagnosing most age-related diseases. Even if we did, it would not be financially possible to run hundreds of diagnostic and pre-screening tests on every healthy adult coming in for an annual check-up.
This proposal seeks to address both of these issues by building a panel of accurate diagnostic tests that can be obtained through a single blood test. This kind of test would be able to diagnose multiple diseases that would often require consultation and authorization from a specialist (which rarely happens before presence of symptoms). The test would also be inexpensive enough that it could be performed on healthy adults on a regular (i.e. annual) basis in order to monitor possible changes in health.
Execution of our proposal relies on new data collected by the UK BioBank (and by our lab) the measures the concentration of thousands of proteins in the blood. Data from our own lab suggests that we can train artificial intelligence to learn how to spot diseases like Alzheimer’s disease just by scanning the concentration of different proteins. The main focus of our proposal will be to train many different artificial intelligence programs to recognize and differentiate many types of diseases. This will be made possible for the first time thanks to the enormous size and high quality of UK BioBank data. We will train the programs such that, when they fail, they are unlikely to falsely signal the presence of the disease when there is none (at the cost of potentially missing some positive cases). Importantly, we will test whether these programs can also predict changes to other known medical markers that might become abnormal before diseases begin to show symptoms.
As a final step, we will test these programs in a real clinical setting to see if they can diagnose diseases, or whether they can forecast future disease in healthy people. This will be an important test to see if a single blood test can predict the presence of multiple diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/building-a-multimodal-imaging-classifier-for-prediction-of-clinical-outcomes-in-brain-related-disorders

Building a multimodal imaging classifier for prediction of clinical outcomes in brain related disorders

Last updated:
ID:
29985
Start date:
15 March 2018
Project status:
Closed
Principal investigator:
Dr Karsten Borgwardt
Lead institution:
ETH Zurich, Switzerland

The aim of this project is to improve the prediction of disease health status using brain imaging data. We benefit from Machine Learning techniques to develop a multimodal classifier working on features extracted from different types of Brain MRI (including anatomical, diffusion and functional). Our primary goal is to build a classifier to predict a phenotype of interest, such as a type of brain disorder or disease prognosis, from multiple imaging modalities. We also aim to make an emphasis on learning interpretable features, allowing to find associations between image patterns and phenotype. Understanding the complexity of our brain is a key factor to facilitate clinical prediction, prevention and treatment of brain disorders, such as Dementia, Alzheimer’s disease, and Unipolar Depression. While it is known that certain biomarkers extracted from specific imaging modalities might be informative for a given disease, we are still far from understanding how we could benefit from efficiently combining multiple imaging modalities and other sources of information. The UK Biobank is an extremely valuable data source for this purpose, providing imaging data obtained with different techniques as well as clinical information on a large scale number of subjects. We will build a machine learning pipeline to perform the following two key steps: (1) learn informative features across multiple imaging modalities and (2) use those features to design improved clinical predictors. Building on state-of-the-art machine learning methods, we will develop an algorithm to predict health-related phenotypes from multiple Brain MRI data and to identify the features that are most associated with a disease and its progression. Upon completion, our research will help clinicians make better use of machine learning to improve diagnosis and personalise medical treatment. Full cohort for brain imaging data and the complementary clinical information.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/building-a-prediction-and-early-warning-system-of-cardiovascular-disease-based-on-artificial-intelligence

Building a prediction and early warning system of cardiovascular disease based on artificial intelligence

Last updated:
ID:
106027
Start date:
8 November 2023
Project status:
Current
Principal investigator:
Professor Xiaoping Li
Lead institution:
University of Electronic Science and Technology of China, China

Cardiovascular disease(CVD), the number one cause of death worldwide,is a group of heart and blood vessel diseases that includes coronary heart disease, cardiomyopathy, arrhythmia, valvular heart disease and congenital heart disease.
In order to obtain the genetic subtypes of CVD with clinical intervention significance, reduce the mortality of CVd and effectively prevent sudden death, in this study, biobank data and data information of CVD patients in hospitals were used to screen candidate pathogenic genes for CVD, and gene mutation data combined with multimodal data were used to classify gene subtypes of CVD and provide clinical significance, multi-center data were used to verify the validity of the prediction model for guiding clinical practice.
Through various methods such as machine learning and deep learning of artificial intelligence, the prediction model and early warning system of death and sudden death of CVD are constructed to identify high-risk patients with clinical death and sudden death, and the fusion model containing pathogenic gene and mutation information is constructed to further improve the accuracy of the model.
This study is helpful for clinical identification of high-risk patients, timely medical intervention and intensive treatment to prevent sudden death, thereby reducing the risk of death and improving the prognosis of patients.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/building-an-online-resource-for-global-investigation-and-validation-of-trans-ethnic-phewas-results

Building an Online Resource for Global Investigation and Validation of Trans-ethnic PheWAS Results

Last updated:
ID:
37539
Start date:
29 March 2020
Project status:
Closed
Principal investigator:
Dr Bingshan Li
Lead institution:
Vanderbilt University, United States of America

Electronic medical records (EMRs) combined with genetic data are a powerful resource for discovering relationships between genes and diseases, and can show which genes are shared between diseases. BioVU, the Vanderbilt DNA biobank, is a rich resource with nearly 250,000 DNA samples linked to EMRs. The Million Veterans Program (MVP) cohort contains EMRs linked to over 400,000 DNA samples. Our goal is to create an online web portal cataloguing all disease-gene relationships in UK Biobank, BioVU, and MVP. Our web portal will serve as a valuable, publicly available resource which may yield unique insights into the genetic factors underlying several diseases, and can help to validate findings from other resources. Our approach has the potential to improve understanding of several diseases including cancer, cardiovascular disease, metabolic dysfunction, neurological disease, and immune system disorders, and opens the door to personalized medicine approaches that may impact clinical diagnosis, treatment, and prevention strategies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/building-and-evaluating-multivariate-statistical-machine-learning-methods-for-knowledge-extraction-from-the-uk-biobank

Building and evaluating multivariate statistical machine learning methods for knowledge extraction from the UK Biobank

Last updated:
ID:
34077
Start date:
24 October 2018
Project status:
Current
Principal investigator:
Professor Thomas Nichols
Lead institution:
University of Oxford, Great Britain

Methods to allow joint analysis across the UK BioBanks’s many imaging, genomic, environmental and clinical variables and remains challenging and underdeveloped. We will develop scalable multivariate statistical machine learning methods and software to extract useful features from all imaging UKBB different data modalities simultaneously to a) to predict different health outcomes from imaging and non-imaging, b) associate brain features with non-brain factors while controlling for individual differences in environmental and genomic data, and c) use UKBB data as a replacement for Monte Carlo simulations in the evaluation and benchmarking of new and existing analyses methods.
Our work will assist scientist in extracting features from the multitude of UK Biobank variables, and finding relationships among these features, ultimately supporting the UK Biobank aims to improve the prevention, diagnosis of disease. We will develop methods and software that uses the shared information among different data modalities in the UKBB to extract features, ultimately building models to predict different health outcomes and associate brain related features with non-imaging variables. We will also use the UK biobank data to benchmark the performance of statistical methods that researchers use everyday.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/building-explainable-multi-omics-prediction-models-for-nash-biomarker-discovery

Building explainable multi-omics prediction models for NASH biomarker discovery

Last updated:
ID:
88396
Start date:
24 May 2022
Project status:
Current
Principal investigator:
Dr Marco Salvatore
Lead institution:
Abzu Ltd., Denmark

Non-Alcoholic Fatty Liver Disease (NAFLD) is currently the most common cause of long-term liver disease worldwide, and more and more people are affected by it every year. As the disease progresses and turns into nonalcoholic steatohepatitis (NASH) the scarring and damage to the liver increases, which has been associated with a heightened risk of liver cancer, heart disease and overall mortality. Most patients do not show any symptoms in the early stages of the disease, which means that often diagnosis happens too late for effective treatment. In addition, accurate diagnosis today is only possible by taking a liver biopsy, which requires an invasive procedure. However, most patients suspected to have NAFLD/NASH are not confirmed. This means a large number of people unnecessarily undergo an invasive procedure and early diagnosis of non-alcoholic steatohepatitis (NASH) is often not possible. Accurate diagnosis of fibrosis risk is crucial for better treatment and management of the disease. In this project we aim to use state-of-the-art interpretable machine-learning technology in combination with one of the richest health datasets in the world to find new biomarker signatures that could help us develop non-invasive tests that could diagnose patients earlier and more accurately than is done today.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/building-machine-learning-models-for-breast-cancer-risk-prediction

Building Machine Learning Models for Breast Cancer Risk Prediction

Last updated:
ID:
41896
Start date:
25 September 2018
Project status:
Current
Principal investigator:
Mr Mahmoud Aldraimli
Lead institution:
University of Westminster, Great Britain

Reducing mortality rates from major NCDs is at the heart of the World Health Organisation (WHO) agenda [1].
The UK NCD profile published by the WHO in 2014 showed that NCDs account for approximately 89% of all mortality [2]. Public Health England published in 2014 that a quarter of the UK population has a long-term medical condition (including major NCDs) and the number of people with multiple conditions is expected to rise [3].
This study aims to identify factors which influence the risk of breast cancer occurrence. This is an important aim as in the long term this could lead to better understanding of the interplay between different illnesses and breast cancer occurrence.
Our approach will be to use Artificial Intelligence (AI) to identify whether there is a link between factors associated with the increased risk of diabetes, obesity and CVD and breast cancer. In particular, we will use a Machine Learning (ML) approach for intelligent data analysis supported by the current digital revolution in collecting and storing data.
Following a systematic review of the UK Biobank, we plan to analyse raw and derived medical and non-medical variables. The analyses will examine whether these variables are correlated with the occurrence of breast cancer, whether these relationships persist in the presence of other variables, and the potential role of obesity, diabetes and CVD in the breast cancer risk prediction.
We will impute missing values while accounting for uncertainty and come up with a predicted risk value of breast cancer. Although the new model will be used to predict breast cancer risk, we will examine our approach for suitability of predicting other NCDs such as obesity, diabetes and CVD. The performance of each model will be assessed mathematically and clinically.
The results may influence a substantial review of our current public health measures to prevent breast cancer.
The research is funded by the Quintin Hogg Trust, the duration of the research is 36 months as part of a PhD programme.

References:
[1] Global Action Plan for the Prevention and Control of NCDs 2013-2020, available at: http://www.who.int/nmh/events/ncd_action_plan/en/ [Last access: 16/04/2018]
[2] World Health Organization, 2014, Non-communicable Diseases (NCD) Country, Profiles. available at: http://www.who.int/nmh/countries/en/ [Last access: 20/04/2018]
[3] K Fenton, Feb 2014, Health and Wellbeing, Reducing the burden of disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/building-polygenic-risk-scores-for-common-complex-traits-in-populations-with-varying-ancestry-background

Building polygenic risk scores for common complex traits in populations with varying ancestry background

Last updated:
ID:
52860
Start date:
11 December 2019
Project status:
Closed
Principal investigator:
Mr Kimmo Aro
Lead institution:
Negen Ltd, Finland

Clinical genetics focuses currently largely to rare genetic syndromes and rare high-risk genetic variants. Novel approaches enable more extensive utilization of genome-wide tools in personalized medicine. These tools could enhance disease risk prediction and preventive patient cascades.

Negen is a commercial company building genetic risk evaluation tools for health care and private consumers. The aim of this research project is to build, test and validate polygenic risk scores (PRS) for various common traits in UK Biobank data. These PRS combine information on genetic and environmental risk factors. The aim is to build PRS at least for the following diseases/traits: coronary heart disease, diabetes types 1 and 2, ischemic stroke, inflammatory bowel disease, asthma, COPD, osteoarthritis, rheumatoid arthritis, breast cancer, colon cancer, prostate cancer, atrial fibrillation, dyslipidemias, deep vein thrombosis and dementias. Validation of any genetic tool is essential before these tools can be utilized in clinical health care. The aim of this project is to validate all genetic tools across different populations. The duration of the project is 3 years.

The validation or novel genetic tools and building the personalized medicine approach has potentially significant public health impact. Implementation of these novel genetic tool in health care may enhance personalized medicine approach, effective risk prediction and treatment, earlier interventions and improved health outcomes.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/building-validating-and-applying-precision-neuroscience-methods-for-mental-health-inference-and-prediction

Building, validating, and applying precision neuroscience methods for mental health inference and prediction

Last updated:
ID:
140089
Start date:
10 May 2024
Project status:
Current
Principal investigator:
Dr Stephanie Noble
Lead institution:
Northeastern University (USA), United States of America

Neuroimaging research has transformed our understanding of mental health. However, recent evidence has brought to light some significant concerns about the rigor and reproducibility of common neuroimaging practices, which undermines the quality of research in the field. This presents a barrier for using this research to develop practical solutions for individuals struggling with mental health issues. One fundamental avenue for addressing these issues is to create and use tools that facilitate higher quality research.

To that end, this project aims to build and validate “precision neuroscience” methods that will enable researchers to 1) conduct more accurate analyses and 2) reveal individual-level insight. Furthermore, this project will use these methods to better understand the complex interaction between brain, body, and environmental factors that underlie mental health outcomes. Importantly, the unprecedented UK Biobank dataset enables us to empirically validate these methods and use them to aid in discovery with the highest available level of accuracy. Overall, this project will result in the creation and application of methods that are expected to improve the overall quality of neuroimaging-based mental health research, leading us closer to real-world utility of this avenue of research.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/built-environment-and-health-in-the-uk-biobank-exploring-the-complex-relationship-using-machine-learning-approaches

Built environment and health in the UK Biobank: exploring the complex relationship using machine learning approaches

Last updated:
ID:
43748
Start date:
25 January 2019
Project status:
Closed
Principal investigator:
Professor Kazem Rahimi
Lead institution:
University of Oxford, Great Britain

This proposed research forms part of a collaborative project led by an interdisciplinary team of researchers with expertise in epidemiology, clinical medicine, machine learning and geospatial information science. We plan to explore how environmental factors, including, but not limited to, physical structures surrounding individuals may influence health and various disease conditions. The UK Biobank collects information about the built environment of the study participants, and we aim to harness the availability of such data resource to examine differences in built environment across the country, and how these differences show similar (or different) patterns of associations with health status, occurrence of diseases, or clustering of disease conditions, across these areas. We will use a range of data science and machine learning approaches to extract, infer, and validate knowledge derived from analysing the data. By analysing the complexity of the relation between built environment and health, we hope to evaluate how well these environmental factors predict various disease conditions as individual factors or as clusters of these factors.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/built-environment-and-obstructive-lung-disease

Built Environment and Obstructive Lung Disease

Last updated:
ID:
26492
Start date:
15 August 2016
Project status:
Current
Principal investigator:
Dr Chinmoy Sarkar
Lead institution:
University of Hong Kong, Hong Kong

This study aims to primarily answer the question: ?where does or does not chronic obstructive lung disease (COPD) and asthma arise, persist and progress in the UK??, describing location in detail through measurable attributes of the built environment in which subjects reside. Secondarily, we aim to generate hypotheses to test regarding the relationship between the built environment and respiratory outcomes. Finally, we aim to use historical and prospective data on patients who meet the case definition of COPD and asthma to test our hypotheses about the effect that the built environment has on patients with COPD and asthma. While asthma can be chronic but reversible, COPD is an irreversible illness affecting an estimated 3 million adults in the UK, with two-thirds of these as yet undiagnosed. We plan to study the relationship of the built environment and respiratory outcomes (COPD & asthma), meeting the UK Biobank?s stated purpose of improving the prevention, diagnosis and treatment of a serious, high-burden disease in the UK. We will draw applications to how urban and environmental choices impact those who suffer from respiratory disease, and assist health professionals to improve diagnosis, counseling and health maintenance for their patients. This study is a collaboration between the University of Hong Kong Faculty of Architecture and School of Public Health. We will take descriptive and analytic approaches, utilizing data from the UK Biobank and the UK Biobank Urban Morphometric Platform (UKBUMP). The UK BUMP is an individual-level built environment database of approximately 750 health-specific built environment metrics for all participants in the UK Biobank prospective cohort. We propose to include the 350,000 participants across 14 UKB collection centres for whom the UKBUMP data will be available by 31st December 2015. Over the duration of the project, data on full cohort will be requested as the complete UKBUMP database becomes available. Once primary care data is also available we will also conduct similar analyses for asthma outcomes.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/burden-of-rare-genetic-variants-in-movement-disorders-and-other-related-neurological-conditions

Burden of rare genetic variants in movement disorders and other related neurological conditions

Last updated:
ID:
60294
Start date:
30 March 2022
Project status:
Current
Principal investigator:
Dr Ryan Yuen
Lead institution:
Hospital for Sick Children, Canada

Movement disorders are common neurological disorders. For example, tics can be found in 4%-19% in children. However, majority of the individuals with movement disorders do not have a genetic cause determined. Our aim is to conduct a genetic study of the children who do not currently have a genetic cause of their movement disorders identified. Genome sequencing technology has allowed detection of causal genetic factors in a subset of these disorders, but the causes in most of the cases are still unknown. This can be due to the neglection of repetitive DNA regions, which constitute most of our genetic information. There are over 50 genetic disorders currently linked to abnormality in repetitive sequence. They are mostly caused by increased length of the repeats (repeat expansions). We will adopt a genetic approach that has been successfully applied on other disorders, such as autism. We will 1) identify and validate tandem repeat expansions in movement disorders, and 2) annotate the identified tandem repeats for possible functional roles in movement disorders. By reviewing these cases and compare them with those in the UK Biobank in the next 3 years, we hope to be able to identify genetic changes in order to find genes that are relevant to movement disorders or their related conditions, and ultimately improve the medical management and treatment for the children.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/burden-of-variants-in-obesity-related-genes

Burden of variants in obesity-related genes

Last updated:
ID:
40415
Start date:
26 October 2018
Project status:
Closed
Principal investigator:
Dr Ida Hatoum Moeller
Lead institution:
Rhythm Pharmaceuticals, Inc., United States of America

The hypothalamic melanocortin 4 receptor (MC4R) pathway plays a critical role in controlling energy expenditure. Alterations in genes within this pathway, including in POMC, LEPR, and PCSK1, have been shown to lead to hyperphagia and severe forms of monogenic obesity. In addition, other genes positioned within this pathway are predicted to contribute to MC4R pathway-mediated obesity. We seek to evaluate UK Biobank data to determine: 1) the association between potentially deleterious variants in the MC4R pathway and obesity, 2) the prevalence of these variants in the UK Biobank population, and 3) the “genetic burden” of carrying multiple variants in different genes within this pathway.

To achieve these aims, we will first identify literature-derived, experimentally tested, and computationally predicted deleterious variants in obesity genes of interest. Next, we will use UK Biobank data to determine the association between these variants and obesity. We will also construct models to determine whether having multiple variants both within and across candidate genes increases the probability of having obesity. Finally, we will estimate the overall prevalence of both individual variants and high-impact genetic burden combinations in the UK Biobank population. We expect this project will take approximately 3 years to complete.

We anticipate that the findings from this study will be relevant to public health in several ways. First, results from these analyses will determine the burden of severe forms of genetic obesity in the UK Biobank population. Second, combinations of deleterious variants may identify new subclasses of obesity for which precision medicine treatments targeting these variants may ultimately be developed. Finally, these results may improve the treatment of serious and life-threatening forms of obesity by identifying the subset of individuals for whom novel therapeutics that target the MC4R pathway may be particularly beneficial.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/caffeine-consumption-postmenopausal-hormone-use-and-breast-cancer-risk-in-postmenopausal-women

Caffeine consumption, postmenopausal hormone use and breast cancer risk in postmenopausal women

Last updated:
ID:
19060
Start date:
1 August 2016
Project status:
Closed
Principal investigator:
Lusine Yaghjyan
Lead institution:
University of Florida, United States of America

Caffeine has been inversely associated with breast cancer risk in some, but not all previous studies. Caffeine and estrogen metabolism pathways share common enzymes and some of these enzymes are induced by caffeine. Postmenopausal hormones (PMH) increase breast cancer risk. Whether there is an interaction between PMH and caffeine with respect to breast cancer risk is unknown. Further, limited data exist on the association of caffeine with breast cancer risk by tumor?s aggressiveness. We will examine interactions of caffeine and PMH with respect to breast cancer risk in postmenopausal women, both overall and by tumor’s aggressiveness Breast cancer remains one of the leading causes of cancer incidence in industrialized countries. Yet, a lot of questions remain open about etiology and prevention of breast cancer as well as risk prediction. The findings of this analysis will help to better understand how these two highly prevalent exposures might jointly contribute to breast cancer risk thus adding to the knowledge in this important area of high public health concern, in line with the UK Biobank purpose. We will identify women who were postmenopausal at the time of enrollment in UKBiobank and did not have a history of breast cancer (BC). Women who developed BC during the follow-up will be compared to women who did not have any cancer (other than non-melanoma skin) with respect to their hormone use and caffeine consumption patterns. We will examine these associations both overall as well as while stratifying tumor’s based on their invasiveness (defined using combination of tumor features including size, grade, nodal involvement, and estrogen receptor status). We will include all women who were postmenopausal at baseline and did not have a history of cancer at enrollment. Women without breast cancer but any other cancer developed during the follow up will be excluded. The study will include all women who meet these criteria


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/calculation-of-a-polygenic-score-for-circadian-depression-its-association-with-clinical-variables-and-predictive-value-for-mood-disorders

Calculation of a polygenic score for circadian depression, its association with clinical variables, and predictive value for mood disorders.

Last updated:
ID:
129030
Start date:
28 February 2024
Project status:
Current
Principal investigator:
Dr Emiliana Tonini
Lead institution:
University of Sydney, Australia

Depression affects more than 300 million people worldwide and is amongst the highest-ranked cause of disability. The cause and biological processes underlying depression remain unclear, while traditional treatments are unsuccessful for many. We hypothesize that a dysregulation of the circadian system, a biological network regulating the timing of all physiology and behavior, plays a causative role on the onset, course, and treatment of a subgroup of depression, referred to as “circadian depression”. Individuals in this subtype expressed features such as delayed sleep-wake phase, hypersomnia, non-restorative sleep, low energy, prolonged fatigue, weight gain and somatic complaints. In this project, we aim to construct a circadian depression phenotype using variables available in the UK Biobank, and to calculate a GWAS for this phenotype to identify genes involved in this pathophysiological pathway. Next, we will calculate a polygenic score of circadian depression to investigate associations between genetic vulnerability for circadian depression and key clinical variables, such as poor response or adverse effect to traditional antidepressant treatments, atypical features, such as weight gain, increased appetite, subjective and objective measures of activity, and chronotype. We will also test whether genetic vulnerability contributes to predicting the onset of mood disorders in young people.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/can-7-day-wrist-worn-accelerometry-predicts-hand-grip-strength-in-healthy-older-adults-and-people-with-multimorbidities

Can 7-day wrist-worn accelerometry predicts hand grip strength in healthy older adults and people with multimorbidities?

Last updated:
ID:
47845
Start date:
7 May 2019
Project status:
Closed
Principal investigator:
Mr Salvatore Tedesco
Lead institution:
University College Cork, Ireland

Wrist-bands are massively diffused nowadays, especially in young and sport-oriented cohorts, but limited adoption is observed in older adults. The main limitations are related to the lack of medical insights that current mainstream wristbands may provide to older subjects. One of the most important aspects investigated by researchers is the possibility to extrapolate clinical information from these wearable devices.
Our research question is to understand if raw accelerometry data collected for 7-days using a consumer-based wristband device, in conjunction with data-driven machine learning algorithms, may be suitable to predict hand grip strength in a number of cohorts: older subjects (> 65 years) with single-morbidities, healthy older adults, and in older adults with multi-morbidities. Morbidities considered in this study are chronic heart failure (CHF), coronary heart disease (CHD), diabetes, and chronic obstructive pulmonary disease (COPD).
The study is not only an ICT/health-related research, but will pave the way to the adoption of wearable devices as an efficient tool for clinical assessment in elderlies with multimorbities, improving and advancing integrated care, diagnosis and early screening of a number of widespread diseases.
This study is supported by EU H2020 funded project ProACT under grant agreement No. 689996. Aspects of this work have been supported in part by a research grant from Science Foundation Ireland (SFI) and is co-funded under the European Regional Development Fund under Grant Number 13/RC/2077. Aspects of this work have been supported in part by INTERREG NPA funded project SenDOC. We foresee that the project will be completed by September 2020.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/can-physical-activity-mitigate-the-decline-in-cognitive-function-associated-with-hearing-loss-a-uk-biobank-analysis

Can physical activity mitigate the decline in cognitive function associated with hearing loss? A UK Biobank Analysis

Last updated:
ID:
628570
Start date:
11 March 2025
Project status:
Current
Principal investigator:
Miss Jessica Rose Andrew
Lead institution:
Lancaster University, Great Britain

Hearing loss (HL) and cognitive decline are major public health concerns, particularly in ageing populations as they reduce quality of life and increase risks of depression and social isolation. Physical exercise (PE) has been shown to mitigate cognitive decline, but its role in hearing health and its interaction with cognitive function is unclear.
This project will analyse UK Biobank data to investigate relationships between hearing ability, cognitive performance, and PE. Key research questions: 1) Is HL associated with cognitive function? 2) Does PE mitigate the relationship between HL and cognitive function? 3) Do demographic and socioeconomic factors moderate these relationships between HL, cognitive function and PE? Objective measures, including Speech-in-Noise hearing test results, cognitive performance metrics, and accelerometer-based activity data, will test our hypotheses: 1) HL will be associated with poorer cognitive performance. 2) Higher levels of PE will reduce the negative impact of HL on cognitive function. 3) Demographic and socioeconomic factors, such as younger age and higher education, moderate these effects.
Variables, such as age, sex, education, and income, will be covariates in statistical models to account for their influence on these relationships. Interaction terms (e.g., Hearing x Education) will test how these variables alter the strength or direction of relationships. Mechanisms like sensory deprivation and cognitive load link HL to cognitive decline, while exercise promotes brain health via improved blood flow, reduced inflammation and neuroplasticity. Existing studies often rely on subjective data, introducing bias.
Using objective measures, this study addresses gaps by examining how PE moderates hearing-cognition links. Leveraging the UK Biobank dataset, findings could guide public health initiatives promoting exercise as a preventative strategy for HL and cognitive decline, with tailored interventions for at-risk populations.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cancer-genetics-variant-interpretation-and-gene-discovery

Cancer Genetics Variant Interpretation and Gene Discovery

Last updated:
ID:
76689
Start date:
4 March 2022
Project status:
Current
Principal investigator:
Professor Clare Turnbull
Lead institution:
Institute of Cancer Research, Great Britain

Identifying people who are inherently at increased risk of developing cancer may lead to better standards of care through enhanced screening and treatment options. We plan to use the genetic data within UK Biobank to look for changes in genes that are more common in people with cancer compared to healthy individuals, assisted by merging the data with various in-house and external genetic and other data. These data will contribute to the activities of and be shared via CanGene-CanVar, a 5 year, 4.3 million Cancer Research UK Catalyst award program.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cancer-incidence-and-premature-mortality-associated-early-phenotypic-risk-markers-related-to-lifestyle-diabetes-mellitus-obesity-other-chronic-diseases-an-uk-biobank-prospective-cohort-study

Cancer Incidence and Premature Mortality Associated Early Phenotypic Risk Markers related to Lifestyle, Diabetes Mellitus, Obesity, other Chronic Diseases: An UK Biobank Prospective Cohort Study.

Last updated:
ID:
48860
Start date:
31 March 2020
Project status:
Current
Principal investigator:
Dr Miguel Angel Luque-Fernandez
Lead institution:
Biomedical Research Institute of Granada, Spain

Cancer is already the second leading cause of death globally and is estimated to account for 9.6 million deaths in 2018, and the global cancer burden is expected to increase by 70% in the coming two decades.

In fact, cancer and other chronic diseases (or non-communicable diseases [NCDs]) share many common risk factors, including aging and unhealthy lifestyles, such as smoking, unhealthy diet, physical inactivity, obesity, and alcohol misuse. Certain NCDs, such as diabetes, may also predispose to cancer independent of these shared risk factors; however, most cancer prevention strategies focus on promotion to adoption of healthy lifestyle and on reduction in exposure to established cancer risk factors. Besides, available research evidence typically focused on the relationship between individual diseases and cancer risk or mortality.

Perceivably, these NCDs often occur hand-in-hand (e.g. diabetes and obesity), we might get a better understanding of their contribution to cancer risk if we can study them at the same time. Because few data are available on this topic, we aim to test the individual and combined effects of type II diabetes mellitus, obesity, markers of metabolic syndrome and other chronic diseases on the risk and mortality of non-Hodgkin lymphoma, breast, prostate, lung and colorectal cancers.

The proposed project will use existing data collected by UK Biobank and will take approximately 58 months to complete.

A better understanding of the biological basis for cancer incidence and mortality would inform cancer prevention and treatment policy and strategies in the future.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cancer-landscape-across-multi-ethnic-populations-integrating-multimodal-data-for-risk-stratification-and-prognostic-insights

Cancer landscape across multi-ethnic populations: integrating multimodal data for risk stratification and prognostic insights

Last updated:
ID:
971764
Start date:
28 September 2025
Project status:
Current
Principal investigator:
Professor Claude Chelala
Lead institution:
Queen Mary University of London, Great Britain

Significant health inequalities exist within ancestry-diverse communities including higher rates of certain cancers compared to the wider population. It is now widely accepted that there are differences in the genetics, tumour biology, and immune environment of certain cancers in ancestry groups. These differences may contribute to differences in prevalence, aggressiveness, and treatment response. Our recent study showed that women of non-white ancestry develop breast cancer earlier, have distinct mutational landscapes, and respond differently to therapies (Nature Communications; PMID:40394000). Our study also suggested that ancestry-specific markers of risk will help support more equitable, population-specific approaches.
Here, we aim to expand our findings for a better understanding of the spectrum of cancers affecting populations from different ancestral background, particularly focusing on non-European ancestries compared to Europeans, through the lens of their inherited risk and lifetime clinical trajectory. For those with cancer, we will derive information on observable demographic, lifestyle and health indicators, and integrate the learning with individuals’ hereditary traits in order to identify ancestry-specific similarities and differences in cancer risk and prognosis. The project is designed to be developed in 3 years and involves initial analysis at pan-cancer level and subsequent analysis of single cancer types with sufficient sample size.
By utilising the repertoire of UK Biobank’s clinical and genomic data, the project ultimately aims to identify key cancer risk and prognostic factors specifically related to the unique demographics of populations from different ancestries.! We expect to contribute towards the benefit of populations at-risk across different ancestry groups and improving cancer outcomes.!


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cancer-risk-and-cancer-outcomes-among-sexual-minorities-compared-with-heterosexual-men-and-women

Cancer risk and cancer outcomes among sexual minorities compared with heterosexual men and women

Last updated:
ID:
42861
Start date:
16 October 2018
Project status:
Closed
Principal investigator:
Dr Catherine Saunders
Lead institution:
University of Cambridge, Great Britain

Aims

The aim of this research is to describe patterns of cancer risks among people from people from sexual minorities and to understand what happens after cancer has been diagnosed.

Scientific rationale

To date, information on cancer risks and cancer outcomes (for example whether someone is diagnosed at an earlier, more treatable stage, or whether their treatment is successful) among people from sexual minorities is limited, despite the existence of some evidence of inequalities. NHS strategy documents focusing on inequalities only include minimal detail on sexual minorities. In addition a leading UK Cancer Charity, MacMillan Cancer Support, have highlighted this lack of data on cancer among sexual minorities, and in the US the American Society for Clinical Oncology have also recently highlighted the need for better understanding of cancer outcomes among sexual minorities. This research will address this need for evidence; it will also allow us to replicate a previous study in an independent data set to improve the scientific rigour of the work that we have already done.

Project duration

We are proposing several linked analyses, starting with a piece of methodological work to understand how best to use sexual behaviour responses in the baseline survey in our further analyses; we therefore expect this analysis to take up to 36 months to complete all phases.

Public health impact

This research will provide evidence to improve the measurement and monitoring of inequalities in cancer risks and outcomes among sexual minorities. It will also support the development of appropriate methodological approaches for researchers and policymakers. Finally the work will support the development of the most appropriate applied approaches to addressing these disparities.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cancer-risk-of-fads1-haplotypes

Cancer risk of FADS1 haplotypes

Last updated:
ID:
43418
Start date:
19 December 2018
Project status:
Current
Principal investigator:
Professor Karsten Suhre
Lead institution:
Weill Cornell Medicine - Qatar, Qatar

Genetic variation not only predisposes to disease, but also influences physiological processes in the human body. Some of these influence how our internal biochemistry works, for example how efficiently our organism can specific nutrients. As an example, genetic variants in a key protein of the human lipid metabolism (FADS1), change levels of omega-3 and omega-6 fatty acids in blood, but also influence the risk of developing cancer. In this project we aim at obtaining a better understanding of these processes at this specific example, especially focusing on how multiple genetic variants may interact in the process of developing cancer. If successful, our work may help mitigate the risk of developing cancer by providing a better understanding of the biochemical process that underlie its development, and to find out whether some fatty acids may be more beneficial to protect the body from disease than others.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cancers-cognitive-and-inflammatory-diseases-patient-survival-using-the-uk-biobank

Cancers, Cognitive and Inflammatory Diseases, Patient Survival Using the UK Biobank

Last updated:
ID:
547729
Start date:
25 June 2025
Project status:
Current
Principal investigator:
Dr Yi-Jhih Huang
Lead institution:
National Defense Medical Center, Taiwan, Province of China

Research Questions:
1. What is the association between primary and second primary cancers and the development of cognitive diseases?
2. How do inflammatory diseases influence patient survival among cancer patients?
3. What are the combined effects of cancer, cognitive decline, and inflammation on patient outcomes?
4. How can we detect early cancer based on the metabolites, biomarkers, sequencing data, and clinical imaging?
Aims:
1. To analyze the relationship between different cancer types and the incidence of cognitive and inflammatory diseases.
2. To evaluate the impact of cognitive and inflammatory conditions on the survival rates of cancer patients.
3. To identify potential biomarkers that could predict patient outcomes or detect early cancers.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cannabis-exposure-and-brain-health-among-middle-aged-and-old-adults

Cannabis exposure and brain health among middle aged and old adults

Last updated:
ID:
65753
Start date:
18 March 2021
Project status:
Closed
Principal investigator:
Dr Galit Weinstein
Lead institution:
University of Haifa, Israel

Due to growing use of medicinal and recreational cannabis among elderly populations, it is important to understand the effect of cannabis use on their brain. It is known that cannabis use may impair cognition when used extensively in adolescence. Yet, how it affects the brain when used in older ages is unknown. Therefore, we will explore the association between history and pattern of cannabis use and cognitive function as well as brain structure as demonstrated by brain imaging. Analyses will be cross-sectional looking at cognitive function and brain MRI measures at one point as well as longitudinal looking at change in outcomes over time. Exploring the associations between cannabis use and brain health in older adults offer an opportunity to observe potential harm or benefit of cannabis use in a population at increased risk for cognitive decline and dementia . The duration of this project is estimated to be 3 years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/capturing-factors-of-diabetes-heterogeneity-using-metabolomic-proteomic-and-genomic-fingerprints

Capturing factors of diabetes heterogeneity using metabolomic, proteomic and genomic fingerprints

Last updated:
ID:
319680
Start date:
20 December 2024
Project status:
Current
Principal investigator:
Professor Robert Wagner
Lead institution:
German Diabetes Center, Germany

Our research aims to understand the different types of adult-onset diabetes and their early stages (prediabetes) by analyzing patterns in blood proteins and metabolites. We want to identify these patterns to better predict who might develop serious complications like heart disease, nerve damage, or kidney failure. By doing so, we hope to create more personalized and effective treatments.
Diabetes is a common condition that affects how the body uses sugar. There are different types of diabetes, mainly type 2 diabetes, which is linked to lifestyle and genetic factors, and type 1 diabetes, which is an autoimmune disease. However, not all people with diabetes experience the disease in the same way; they have different risks for complications and different responses to treatment. Recent studies have shown that there are various subtypes of diabetes, each with unique biological characteristics. Understanding these subtypes can help in predicting complications and tailoring treatments more precisely.
The project will take approximately 36 months to complete.
This research has the potential to significantly improve public health. By identifying the specific subtypes of diabetes and their associated risks, we can develop better strategies for preventing and managing complications. This could lead to more effective treatments, reducing the burden of diabetes-related health issues. In the long run, this means healthier lives for people with diabetes and a reduction in healthcare costs associated with managing the disease and its complications.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/capturing-genotype-to-phenotype-relationships-for-complex-phenotypes-using-neural-network-based-statistical-models-that-leverage-phenotype-phenotype-relationships

Capturing genotype-to-phenotype relationships for complex phenotypes using neural network based statistical models that leverage phenotype-phenotype relationships.

Last updated:
ID:
103445
Start date:
25 January 2024
Project status:
Current
Principal investigator:
Dr David Gavin Mets
Lead institution:
Arcadia Science LLC, United States of America

Genetic data is more available than ever before. Consistently, we now see many studies relating genetic factors to human traits. However, these studies frequently only predict poorly. The methodology often used to relate genes and traits does not allow for the possibility of complex interactions between genes, other genes, and environments to impact traits. Here we aim to test our new neural-network-based framework for creating such genotype-to-trait maps. Our approach allows for interactions between both genes and environments and captures correlations among phenotypes. If our method produces more accurate predictions of traits than conventional analysis, we will determine the fraction of that prediction increase that is attributable to gene-gene, potentially gene-environment, and dominance effects. An understanding of the prevalence and importance of these phenomena in determining human traits will improve our understanding of the basic drivers of human trait variation.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/capturing-the-local-environment-and-its-relationship-with-health-outcomes-via-geo-localized-information-extracted-from-satellite-and-map-services

Capturing the local environment and its relationship with health outcomes via geo-localized information extracted from satellite and map services

Last updated:
ID:
60481
Start date:
13 July 2020
Project status:
Closed
Principal investigator:
Mr Andrea Ganna
Lead institution:
University of Helsinki, Finland

The local environment surrounding an individual’s household is an important predictor of socio-economic status and adverse health events. An increasing number of geo-localized information is being collected, e.g. by Google map services. It is, however, unclear how such a large amount of information can be leveraged to explore the effect of the environment on health. Previous studies have shown that information extracted from Google street view can provide an estimate of neighborhood socio-economic level. The goal of this project is to extract from satellite and map services novel environmental features and test their association with health outcomes above and beyond simpler measures of environmental exposure and socio-economic status. To extract novel local environment features we will use deep learning methods applied to satellite and street view images as well as more traditional statistical models. This 5-year project will provide a better understanding of how the local environment can impact an individual’s health and can inform preventive strategies aimed to reduce environmental risk factors.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cardia-cardiovascular-risk-detection-among-women

CARDIA: Cardiovascular Risk Detection among Women

Last updated:
ID:
310110
Start date:
5 November 2024
Project status:
Current
Principal investigator:
Dr Samira Rahimi
Lead institution:
McGill University, Canada

Cardiovascular diseases (CVD) in women and minority groups are not studied enough, not recognized early, not diagnosed well and not treated properly. CVD, including heart failure, heart attack and stroke, is a major global health problem and the leading cause of death among women. CVD killed nearly 18 million people in 2019, and it is estimated that this figure will rise to 22 million by 2030.

Given the insufficient research, recognition, diagnosis and treatment of women at risk of or with CVD, the aim of our project, CARDIA, is to address a gap in CVD prevention. We will develop a new AI algorithm to personalize CVD prevention interventions based on a variety of datasets including the UK Biobank dataset, using an ML methods such as naive Bayes, support vector machines, random forests and neural networks among others. The algorithm will be able to classify patients into different risk categories. Ensemble models, another machine learning method, will also be developed to improve predictive accuracy for capturing different risk factors (e.g., mental health symptoms) more prevalent in women. Additionally, explainable methods (SHAP, LIME, PIMP or Explainable Boosting Machine (EBM)) will be integrated to understand the reasoning behind the algorithm’s predictions. The outcome of our project will be a tool for primary care providers and patients to predict and prevent CVD.

Therefore, CARDIA could: i) address gender disparities in the existing literature, ii) provide technology for the prediction and prevention of CVD based on signs of CVD and iii) impact other prediction and prevention strategies to open new avenues for discovery across various fields.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cardiac-analysis-for-vascular-estimation-cave

Cardiac Analysis for Vascular Estimation (CAVE)

Last updated:
ID:
57170
Start date:
11 March 2020
Project status:
Current
Principal investigator:
Dr Chris John Crockford
Lead institution:
Relative Health Limited, Great Britain

Having built an artificially intelligent machine learning algorithm that can determine Blood Pressure from two physiological standard biomarkers, we wish to test out model on the population Blood Pressure and ECG data held within biobank. Our aims are to identify new biomarkers within the ECG signal that relate to the Blood Pressure directly.

The scientific rationale for doing these works is to establish new linkages between the cardio and vascular side of the cardiovascular system and to provide a new means to acquire Blood Pressure data without having to use time consuming, uncomfortable inflatable cuffs.

The project duration is 6 months.

The public health impact is significant as Blood Pressure is one of the least adhered to physiological parameters.

In the EU nearly 24 % of the deaths of 1.7m persons younger than 75 could have been avoided.
The WHO accredits 63% of global deaths to non-communicable diseases that are largely preventable.
Hypertension – Blood pressure (BP) increases the risk of cardiovascular diseases, strokes, and arterial stiffness.
The Lancet concurs that: Despite the widespread availability of effective treatment, control of hypertension in the community remains sub-optimal. Key reasons cited are clinical inertia, poor adherence, and organisational failure. However it is known that self-monitoring is an effective way to improve BP control. NHS England state cardiovascular disease affects 6 million people and costs £7 billion a year. CAVE addresses this healthcare need by developing novel algorithms to derive BP from ECG signals alone.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cardiac-modeling-for-myocardial-infarction-drug-testing

Cardiac Modeling for Myocardial Infarction Drug Testing

Last updated:
ID:
603483
Start date:
4 February 2025
Project status:
Current
Principal investigator:
Mr Minye Shao
Lead institution:
Durham University, Great Britain

This study aims to leverage digital twin technology for simulating drug interventions in myocardial infarction (MI) patients. By integrating genetic, clinical, and imaging data from the UK Biobank, we seek to develop personalized cardiac models capable of accurately predicting individual cardiac responses to various drugs. The objectives of this research include:
Investigating how genetic and environmental factors influence drug responses in MI patients.
Validating the feasibility of digital twin technology in cardiovascular drug testing.
Providing potential personalized therapeutic strategies to support clinical decision-making.
The scientific rationale of this research lies in employing innovative digital modeling to bridge the gap between generalized clinical trials and individual patient care.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cardio-metabolic-risk-body-composition-and-cardio-metabolic-events-in-women-with-polycystic-ovary-syndrome-pcos

Cardio-metabolic risk, body composition and cardio-metabolic events in women with Polycystic Ovary Syndrome (PCOS)

Last updated:
ID:
74170
Start date:
12 January 2022
Project status:
Current
Principal investigator:
Professor Daniel Cuthbertson
Lead institution:
University of Liverpool, Great Britain

Polycystic ovary syndrome (PCOS) is a common disease which is linked with multiple health conditions including type 2 diabetes, high blood pressure, liver disease and heart disease. By using UK biobank data this project aims to assess the key mechanisms whereby PCOS may contribute to serious health conditions. This will be achieved by assessing body fat distribution via imaging, blood markers of health, exercise level via activity monitors and heart function via ultrasound. Previous imaging studies have demonstrated that in women with PCOS abdominal fat is important in the development of diabetes and heart disease. These studies were small and, in most cases, did not use MRI which is the best measure of body composition. UK biobank presents a unique opportunity to assess the relationship between body composition and PCOS. This may help identify new ways by which the disease develops and progresses to more serious conditions. Assessment of exercise data will allow us to understand whether women with PCOS are less active and if this relates to long term health. Measurement of common blood markers and assessment of liver scans will help delineate whether women with PCOS have poorer liver health. Finally, assessment of the development of heart attacks and strokes will help us understand whether women with PCOS are at greater risk of developing these serious health conditions. Overall, this study is vital as there is currently no large-scale data assessing the relationship between PCOS and these markers of health.

This project will last from August 2021 until ~ August 2022.

This project has considerable potential to improve PCOS prevention and treatment. By identifying the diseases associated with PCOS this will allow for updated public health strategies in the prevention of the disease. For instance, if we associate physical activity level with the development of heart attacks, this will help inform the importance of staying physically active to protect heart health in women with PCOS. Additionally, by identifying diseases closely associated with PCOS this may help in the development of new therapeutic measures to combat the disease’s long-term health consequences.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cardio-metabolic-traits-and-direct-and-indirect-consequences-of-covid-19

Cardio-metabolic traits and direct and indirect consequences of Covid-19

Last updated:
ID:
70676
Start date:
30 June 2021
Project status:
Closed
Principal investigator:
Dr Louisa Gnatiuc Friedrichs
Lead institution:
University of Oxford, Great Britain

Covid-19 is a health emergency that is more harmful among people with pre-existing chronic disease such as diabetes, vascular disease or cancer, and accounts for important mortality, which would not otherwise occur in the absence of a Covid-19 infection. This study aims to carefully characterise what makes some people more likely to get the Covid-19 infection than other people, and what the immediate and long-term consequences of Covid-19 are. The data from the UK Biobank will be primarily used to answer these questions. For comparison, information from the Mexico City Prospective Study might be also assessed. This would be interesting, because in Mexico, more people suffer from diabetes and its complications compared to the UK. Analyses will take 3 to 5 years to conduct and publish, and will ultimately help to better understanding this new disease, why some people are more affected by it compared to others, and its impact among those without and with per-existing diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cardiolifespan-dynamics-harnessing-longitudinal-insights-for-optimal-cardiovascular-health-environmental-risk-factors-and-aging-across-life-stages

CardioLifeSpan Dynamics: Harnessing Longitudinal Insights for Optimal Cardiovascular Health, Environmental Risk Factors and Aging Across Life Stages

Last updated:
ID:
295277
Start date:
17 April 2025
Project status:
Current
Principal investigator:
Dr Karim Lekadir
Lead institution:
University of Barcelona, Spain

The BCN-AIM research projects aim to understand how environmental factors, lifestyle choices, and genetic predispositions interact to affect health throughout life, focusing on major health issues like obesity, type 2 diabetes, cardiovascular diseases (CVD), and aging-related health problems.

Primary goals include developing advanced AI models to improve early diagnosis and prognosis of CVDs, the leading cause of death worldwide, accounting for about one-third of global deaths. Another aim is to create personalized healthcare solutions by analyzing diverse data sources, including medical imaging, ECG data, lifestyle habits, and medication histories.

These projects are scientifically grounded in the complexity and variability of CVDs and chronic conditions. Conditions like heart failure have diverse causes, symptoms, and progression patterns, making them challenging to diagnose and treat effectively. AI models can analyze large datasets to identify patterns and risk factors not evident through traditional methods. For example, AI can detect subtle changes in ECG data indicating early stages of heart disease, enabling timely intervention. Additionally, the projects will adopt a life-course approach, using longitudinal data to study how various exposures and behaviors over time influence health outcomes. This comprehensive approach is essential for understanding the root causes of diseases and developing prevention strategies.

The public health impact of BCN-AIM projects is expected to be substantial. By improving the accuracy and timeliness of CVD diagnosis and prognosis, the projects aim to enhance patient outcomes and reduce mortality rates. Personalized healthcare solutions, developed through AI analysis of comprehensive data, promise to tailor treatments to individual patients’ needs, optimizing clinical outcomes and improving quality of life. Additionally, these projects seek to inform and transform health policies and practices by providing deeper insights into disease prevention and health promotion. Integrating AI and thorough data analysis, the BCN-AIM projects aim to drive significant improvements in public health, support the development of more effective and personalized healthcare solutions, and contribute to the advancement of healthcare delivery systems within European and low-resource regions.

The project duration is flexible due to the iterative nature of generating, testing, and refining hypotheses. The initial 3-year plan (36 months) is designed to validate initial hypotheses, discard those that fail, and develop robust models. This timeframe also allows for ample opportunity to produce and publish findings, ensuring a comprehensive and thorough research process. Annual reports will detail the evolution of research lines, document any deviations, and outline both discarded and successful hypotheses.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cardiomarker-computational-imaging-phenomics-in-population-cardiac-mri-with-automatic-image-quality-assessment-benchmarking-scalability-and-inference-with-state-of-the-art-algorithms

CARDIOMARKER- Computational imaging phenomics in population cardiac MRI with automatic image quality assessment: benchmarking, scalability and inference with state-of-the-art algorithms.

Last updated:
ID:
11350
Start date:
17 January 2019
Project status:
Current
Principal investigator:
Professor Alejandro F Frangi
Lead institution:
University of Manchester, Great Britain

Several cardiovascular conditions like heart failure, coronary artery disease, diabetes, and structural heart disease manifest in alterations of the anatomy or deformation of the myocardium. The hypothesis of this study is that existing tools for cardiac image analysis providing information on 3D cardiac morphology and deformation, developed for small patient cohorts, scale up to handle datasets in the order of hundreds and thousands of subjects. We will simultaneously undertake benchmarking of competing algorithms as demonstrate the impact of image analysis errors on relevant associative and causal inference tasks.
CARDIOMARKER will carry out a large-scale scalability testing of current image analysis tools ultimately helping UK Biobank researchers in extracting objective imaging phenotypic biomarkers of cardiac morphology and deformation correlated to disease presence, severity or progression. We will manually assess a subset of the datasets by two operators in two independent sessions. We will compare the manually segmented cardiac structures (MR cine) and tag intersections (MR tagging) from manual analysis against those of our automatic techniques.
CARDIOMARKER will elucidate how errors in CMR biomarkers influence the strength of associative and causal models. In collaboration with our clinical experts, we will formulate illustrative hypothesis re the association between cardiac morphology/deformation and genetic, lifestyle (activity, body mass composition), metabolism-related (bone ageing, liver function), environmental (exposures), and physiological (HR, BP) variables. We will generate associative/causative models, and will study the influence on those models of errors in CMR biomarkers derived from automatic analysis. This will shed light on the strength of the associations/causal relationships as a function of the size of the population and the noise level in the markers themselves.
We want to answer these questions in relationship to manual delineation and previous performance of the techniques:
a) What is the accuracy in cardiac anatomy delineation in population imaging studies?
b) What is the accuracy of extracted cardiac deformation fields?
c) What is the failure rate of automated methods operating on large-scale population imaging?
d) What is the impact of automated segmentation/registration errors on associative/causative models of phenotype-genotype relationship?
We will deliver the UKBB automatic and objective quantitative imaging information on the full cohort of patients imaged with CMR helping. These will establish population ranges of normality and thresholds of abnormality useful in cardiology. We will deliver knowledge on how to interpret and how errors compound in statistical modelling when attempting to unravel associative/causal relationships involving not only CMR but also other biomarkers.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cardiometabolic-and-genetic-determinants-of-brain-structure-and-function-a-focus-on-obesity-type-2-diabetes-and-associated-factors

Cardiometabolic and genetic determinants of brain structure and function – a focus on obesity, type 2 diabetes and associated factors

Last updated:
ID:
24954
Start date:
13 March 2018
Project status:
Current
Principal investigator:
Professor Velandai Srikanth
Lead institution:
Monash University, Australia

This study aims to study the relative contribution of obesity, cardiometabolic factors and genetic factors to the development of brain dysfunction. The emphasis is on the role of obesity, prediabetes, type 2 diabetes, and their associated factors as exposures. Common factors predisposing to metabolic dysfunction and dementia will be of main interest. The outcomes will be structural and functional measures (cognitive and imaging. The role of sex/gender in modifying these relationships will be evaluated This body of research will shed new light on mechanisms underlying the associations between two highly prevalent disorders (diabetes and dementia), both major public health problems It relates to the major public health disorders of obesity, diabetes and dementia – hence is health-related and in the public interest. Our group has substantial expertise in the field of ageing, advanced imaging analysis, dementia and cognitive dysfunction related to diabetes. Data from the UK biobank will be collated for exposure and confounding/modifying variables. Imaging analyses will be conducted using both existing processed data and with in-house methods to generate outcome variables. Associations of interest will be examined using a combination of simple and advanced analytical methods.

Data arising from these analyses will be presented at scientific conferences, and published in peer-reviewed literature. Given that we are aiming to assess the magnitude of relative contributions of the exposure factors, to explore effect modification and interaction, and to use advanced analytical techniques (including graph/network analyses) – this would require a substantial dataset given the levels of stratification required. The primary dataset includes all people with measured cognitive function, brain imaging, genomics and body composition. Available data on brain imaging in this sample will also be required. Additionally,data are required on relevant demographic, psychosocial, lifestyle,c ardiovascular, genes (SNPs), and already available blood biomarkers collected and analysed in the UK Biobank – reflecting factors relevant to linking obesity, diabetes and dementia.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cardiometabolic-and-lifestyle-determinants-of-cardiovascular-disease-risk-in-breast-cancer-survivors

Cardiometabolic and lifestyle determinants of cardiovascular disease risk in breast cancer survivors

Last updated:
ID:
83076
Start date:
9 March 2022
Project status:
Current
Principal investigator:
Ms Julia Rickard
Lead institution:
University of Toronto, Canada

Breast cancer is the most commonly diagnosed cancer in women in the world. There have been tremendous improvements in early detection and treatment of breast cancer and early-stage breast cancer mortality rates have decreased by nearly 50% in the past 40 years as a result. However, now that more women are surviving their breast cancer diagnoses, there has been a rise in cardiovascular disease in this population. In fact, women with a breast cancer diagnosis are more at risk for cardiovascular disease at all time points following their diagnosis when compared to women without breast cancer. This elevated risk is credited to a variety of factors including cardiotoxic (causes damage to the heart) cancer treatment, poor lifestyle behaviours (e.g. physical inactivity and smoking), the overlap between the risk factors for breast cancer and cardiovascular disease, and ectopic (abnormal location) fat deposition. Our research group is interested in further understanding cardiometabolic dysfunction in breast cancer survivors and its role in elevated cardiovascular disease risk.
This 6-month project aims to evaluate the influence of a breast cancer diagnosis on cardiovascular and metabolic health. Established cardiometabolic risk factors such as blood pressure and lipid profile, cardiac structure and function, ectopic fat volumes, fitness, components of the metabolic syndrome, lifestyle behaviours (such as physical activity and alcohol consumption) and Framingham risk score will be compared between women with and without a history of breast cancer. We also aim to examine the association between lifestyle behaviours and cardiovascular disease risk in all women, and if this risk differs between those with and without a history of breast cancer. In addition, we aim to explore whether menopausal status and the female sex hormone, estrogen, play a role in these relationships.
By establishing the reasons behind the elevated cardiovascular disease risk, we are able to better understand the cardiometabolic dysfunction experienced by breast cancer survivors. This allows researchers to develop risk reduction interventions and strategies to mitigate this risk and improve the health of breast cancer survivors.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cardiometabolic-polygenic-risk-scores-and-its-impact-on-renal-endocrine-sensorial-traits-and-malignancy

Cardiometabolic polygenic risk scores and its impact on renal, endocrine, sensorial traits and malignancy

Last updated:
ID:
77452
Start date:
22 July 2022
Project status:
Current
Principal investigator:
Dr Jacob Shujui Hsu
Lead institution:
National Taiwan University, Taiwan, Province of China

We aim to construct polygenic risk scores (PRS) derived from UK Biobank and Taiwan Biobank to investigate the cardiometabolic risk factors on heart functions, endocrine functions, kidney functions, osteoporosis, and sensory system. A “polygenic risk score” is a way through which individuals can learn about their risk of developing a disease according to the total changes in their genome. We intend to generate PRS by using the genomic data from the UK Biobank and Taiwan Biobank and performing statistical analysis to discover the causal relationship between certain cardiometabolic factors and organ functions within 3 years. The created PRS would improve current healthcare quality in several ways. First, clinical workers can advise people with higher PRS to pay additional attention to certain diseases. In addition, people may receive early healthcare management based on their risk scores. Furthermore, PRS can provide disease insights for medical researchers.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cardiometabolic-risk-factors-and-their-associations-across-the-spectrum-of-cardiovascular-disease-presentations

Cardiometabolic risk factors and their associations across the spectrum of cardiovascular disease presentations.

Last updated:
ID:
794884
Start date:
3 July 2025
Project status:
Current
Principal investigator:
Dr Nathalie Conrad
Lead institution:
University of Oxford, Great Britain

Background: The role of metabolic risk factors in the development atherosclerotic heart disease is well established. Many risk factors are also implicated in a range of other cardiovascular conditions, yet their exact role, effect size, and interactions between them, are less well established.
Objectives: To i) examine and compare the association between different cardiovascular markers and different cardiovascular presentations; ii) identify novel risk factors implicated in the development of cardiovascular diseases, particularly for less commonly studied conditions; iii) examine possible underlying biological mechanisms, by investigating how blood or imaging biomarkers mediate observed associations; iv) develop a novel risk prediction models that considers and balances risks across different cardiovascular presentations.
Methods: The study will use one of the largest and richest datasets available with cardiovascular risk factors, blood biomarkers and imaging data, the UK Biobank. Analyses will consider all variables available in the cohort, investigate risks separately for men and women, and consider 10 cardiovascular outcomes, including acute coronary syndrome; aortic aneurysm; aortic stenosis; atrial fibrillation and flutter; chronic ischaemic heart disease; heart failure; peripheral arterial disease; second- and third-degree heart block; stroke; and venous thromboembolism.
Expected outcomes: A better understanding of possible pathophysiological mechanisms underlying cardiovascular disease across its spectrum of presentations, may support the future development of preventive strategies, treatments and tools for identification of individuals at highest risk of developing cardiovascular events.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cardiometabolic-trait-genetics

Cardiometabolic trait genetics

Last updated:
ID:
10205
Start date:
1 October 2015
Project status:
Current
Principal investigator:
Professor Eleftheria Zeggini
Lead institution:
Helmholtz Zentrum Munchen, Germany

We are requesting access to the full UK Biobank genotype data along with phenotype data on traits of cardiometabolic relevance. The data will be used in genotype-phenotype association studies and will contribute to genome-wide discovery efforts, as well as to replication efforts for already-identified promising signals in independent samples. Our team has expertise in running genetic association studies and we have a primary focus on traits of cardiometabolic relevance. The proposed work fits in with the aims of UK Biobank, as it seeks to identify associations between sequence variants and traits of medical relevance. It leverages the large sample size of the UK Biobank cohort alongside further large-scale genetic studies of the same traits. The focus of our work is on complex trait genetics. We design and carry out large-scale genetic association studies and aim to identify genetic loci associated with traits of medical relevance. We will link genotype to phenotype information to find genetic loci predisposing to human disease and relevant traits. Full cohort


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cardiorenal-genomics-for-risk-prediction-in-african-descent-populations

Cardiorenal Genomics for Risk Prediction in African Descent Populations

Last updated:
ID:
63682
Start date:
25 January 2021
Project status:
Current
Principal investigator:
Dr Nicole Marie Davis Armstrong
Lead institution:
University of Alabama at Birmingham, United States of America

Hypertension (HTN) and chronic kidney disease (CKD) are major risk factors for cardiovascular disease outcomes which disproportionately affect African Americans (AAs). AAs have a higher prevalence of HTN and CKD (40%, 18%, respectively) than EAs (28%, 13%, respectively) and Hispanics (26%, 15%, respectively), and are also more likely to be taking antihypertensives (AHT), yet are less likely to have their blood pressure (BP) controlled. The reasons for these differences are known to include both environmental and inherited factors. To date, environmental contributors are better understood, yet few clinically impactful genetic-risk contributors overburdening AAs have been identified. Some examples include apolipoprotein L1 (APOL1; risk variants G1 and G2 confer up to a 7-fold increased risk for end-stage renal disease), Sickle Cell Trait (HBB rs344 associated with a 2-fold increased risk for end-stage renal disease), and TTR V122I (associated with 2 fold higher odds for heart failure). Overall, more research is needed to continue to characterize genetic risk factors that can help explain and ultimately prevent these disparities. Unfortunately, populations of African ancestry have been severely underrepresented in genetic epidemiology and pharmacogenetic research (AAs represent 400K EAs, <15K AAs). Overall, this research will aid in filling in major gaps in genetic research in this ancestry group. We expect replication efforts from Aims 1-2 to be complete in the next 2 years and for aim 3 in the year 3.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cardiovascular-and-kidney-health-evaluation-cake-advancing-the-knowledge-of-cardiovascular-kidney-metabolic-health

Cardiovascular and Kidney Health Evaluation (CaKE): Advancing the Knowledge of Cardiovascular-kidney-metabolic Health

Last updated:
ID:
429573
Start date:
12 December 2024
Project status:
Current
Principal investigator:
Professor Tazeen Jafar
Lead institution:
Duke-NUS Medical School, Singapore

Aims
This research project aims to advance our understanding of cardiovascular-kidney-metabolic (CKM) health. We will investigate how cardiovascular health, psychological well-being, and high adherence to an environmentally friendly diet contribute to cardiovascular and kidney diseases. Additionally, we will evaluate a new cardiovascular risk prediction model inclusive of kidney function indicators and compare it with conventional cardiovascular risk prediction models.

Scientific Rationale
CKM health is a holistic concept recognizing the interconnectedness of cardiovascular, kidney, and metabolic functions. Chronic kidney disease (CKD) and cardiovascular disease (CVD) often coexist, which complicates the prevention and treatment of these conditions. Recent research suggests that maintaining good cardiovascular health, adhering to a balanced healthy diet, and ensuring psychological well-being can lower the risk of CVD while research on their effects on kidney health remains understudied. Our study seeks to leverage the extensive data from the UK Biobank, which includes health information from over 500,000 participants, to provide more insights on CKM health to bridge the research gaps.

Project Duration
The project is expected to last three years, covering data analysis, interpretation, and dissemination of findings.

Public Health Impact
The results of this research could have significant implications for public health. By advancing our understanding of CKM health, we can develop more effective strategies for preventing and managing chronic diseases, leading to better health outcomes and quality of life. Research findings on the effects of maintaining optimal cardiovascular health on adverse kidney outcomes can help inform holistic prevention and treatment strategies to improve prognosis and quality of life among patients with impaired kidney function. Moreover, this research will provide evidence of the benefits of the EAT-Lancet planetary health diet, which promotes human health and supports environmental sustainability. These findings could inform dietary guidelines while adding knowledge of its benefits of enhancing CKM health. By evaluating and comparing various cardiovascular risk scores, including the newly developed PREVENT equation, the study aims to enhance risk prediction for CVD in different populations. This can help healthcare providers identify high-risk individuals and tailor their treatment plans more effectively. Last, the study will explore the impact of mental health on CKM health, highlighting the importance of psychological well-being in managing chronic diseases and this could lead to integrated care approaches that address both physical and mental health needs.

Overall, our findings will contribute to public health strategies aimed at improving CKM health, ultimately enhancing patient outcomes and promoting a healthier population.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cardiovascular-comorbidities-in-patients-with-psoriatic-arthritis

Cardiovascular Comorbidities in patients with psoriatic arthritis

Last updated:
ID:
171915
Start date:
13 May 2024
Project status:
Current
Principal investigator:
Professor Laura Claire Coates
Lead institution:
University of Oxford, Great Britain

Psoriatic arthritis (PsA) is a type of arthritis associated with psoriasis. Patients with PsA are at higher risk of developing cardiovascular diseases, including heart failure. Heart failure is a condition where the heart is unable to pump blood effectively, and it is a significant cause of morbidity and mortality. PsA patients have a 32% increased risk of developing heart failure compared to the general population. Heart failure can be divided into two types based on whether the left ventricular ejection fraction (LVEF) is reduced or preserved. Some evidence suggests that rheumatoid arthritis may predispose to heart failure with preserved ejection fraction, but data on PsA and heart failure with preserved ejection fraction are lacking. Therefore, there is an urgent need to improve primary and secondary prevention of cardiovascular disease in PsA patients. Abdominal fat deposits, including visceral adipose tissue (VAT), are associated with a higher mortality risk and a higher risk of coronary artery disease. Data on VAT in PsA are lacking, and contradictory results have been published. Furthermore, data on hepatic fat as a marker of metabolic syndrome in PsA patients are lacking. Sarcopenia, defined as a decline in muscle strength and size, has been associated with cardiovascular diseases.
This study aims to compare the abdominal ectopic fat and muscle and their relationship with cardiovascular diseases in patients with PsA, psoriasis, and healthy controls. It is expected that there will be an increase in abdominal fat deposits and sarcopenia in PsA patients compared to healthy controls and patients with psoriasis. The results could help identify patients at high risk of cardiovascular diseases. This study also aims to compare heart failure with preserved ejection fractions in patients with PsA, psoriasis, and healthy controls. If an increase in heart failure with preserved ejection fraction is confirmed, it could raise awareness among clinicians to seek this condition in patients complaining of dyspnea.
This project should last one year.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cardiovascular-damage-in-renal-disease-investigating-factors-linked-to-cardio-renal-disease-constellations-in-the-uk-biobank

Cardiovascular damage in renal disease: investigating factors linked to cardio-renal disease constellations in the UK Biobank

Last updated:
ID:
343445
Start date:
7 October 2024
Project status:
Current
Principal investigator:
Dr Janis Marc Nolde
Lead institution:
University of Freiburg, Germany

Our research project aims to unlock deeper insights into how diseases affecting the kidneys, like chronic kidney disease, hypertension, and diabetes, can lead to heart and blood vessel damage. Using data from the UK Biobank, we will investigate how changes in kidney function and blood pressure over time contribute to cardiovascular health problems.

The connection between kidney health and cardiovascular disease is well-documented but complex. High blood pressure, which affects more than a billion people worldwide and is a leading cause of death, is known to be a major cardiovascular risk factor. However, the nuances of how blood pressure changes with age and how this specifically impacts cardiac health are not completely understood. Similarly, kidney function typically declines with age, influenced by a variety of heart-related risk factors. Through the UK Biobank, we can study these dynamics in detail, examining the longitudinal changes and their impacts on cardiac health.

Our study will make use of advanced statistical and data science techniques, including regression modelling, clustering, imputation for missing data, and potentially machine learning for predictive modelling. This combination of analytical approaches will allow us to handle and interpret the complex and large-scale data provided by the Biobank effectively.

We anticipate that this research will span several years (likely about 3 years), given the scope and depth of the analysis required. Throughout the duration of the project, we will focus on the entire cohort available in the UK Biobank, utilising a range of data types. This includes basic health information, detailed data derived off imaging data that can show signs of heart and blood vessel damage, and results from biological sample assays.

By improving our understanding of how renal diseases influence cardiac health, we can enhance the detection and treatment of cardiovascular issues in patients with kidney disease. This has the potential to lead to earlier and more accurate diagnosis, optimized targeted therapies, and overall improved health outcomes. Such advancements could not only save lives but also reduce the burden on healthcare systems. Ultimately, our findings may lead to improved screening and preventive measures, significantly improving public health in the face of growing rates of both kidney disease and cardiovascular conditions globally.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cardiovascular-event-prediction-using-retinal-images-patient-physiological-information

Cardiovascular Event Prediction Using Retinal Images & Patient Physiological Information

Last updated:
ID:
77343
Start date:
11 October 2021
Project status:
Current
Principal investigator:
Mr David George Prichard
Lead institution:
Reti Health Limited, Great Britain

The aim of this project is to build a tool to identify people at high risk of heart attack and stroke using images of their retina.

The retina is a layer of tissue at the back of the eye which provides a convenient window into vascular health, as it is easy to image, and clearly shows small blood vessels. Previous research using UK Biobank data has shown that it is possible to use retinal images to predict an individual’s likelihood of suffering a heart attack or stroke. We will build on this research to make a product that informs people of their risk when they go for routine eye checks at optometrists.

This has the potential to have a massive impact on public health. Hundreds of thousands of heart attacks and strokes occur in the UK every year, making them the second most common cause of death and costing the NHS £7 billion annually. The majority of these are preventable with early treatment and lifestyle changes, yet millions of people are unaware of their risk. Deployment in an optometry setting would allow cheap, fast screening of millions of people every year, enabling earlier intervention and help stop thousands of deaths and disabilities annually while reducing the burden on the NHS.

The project will take place over 36 months, and involve evaluating the efficacy of the screening method on UK Biobank as well as external datasets to ensure suitability for deployment in an optometry context.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cardiovascular-genetic-risk-score-in-patients-positive-for-sars-cov-2-infection-risk-stratification-cargencors-validation-of-the-significant-single-nucleotide-polymorphisms

CARdiovascular GENetic risk score in patients positive for SARS-CoV-2 infection Risk Stratification (CARGENCORS): validation of the significant single nucleotide polymorphisms.

Last updated:
ID:
84905
Start date:
10 August 2022
Project status:
Current
Principal investigator:
Professor Irene Degano
Lead institution:
Universitat de Vic - Universitat Central de Catalunya, Spain

We will analyze the association between genetic variants and severe coronavirus disease (COVID-19). The genetic variants under study are changes of one letter in the genome. These changes can be associated with diseases such as COVID-19.
We will select COVID-19 patients who had a severe/fatal course of the disease as well as patients who had only mild/moderate symptoms. We will examine 86 genetic variants and the clinical history of these patients to analyze whether there is an association between each of the genetic variants and having severe COVID-19. The 86 genetic variants are present in or near to genes linked with cardiovascular diseases, inflammation and response to infections. We selected these variants because COVID-19 patients with cardiovascular diseases have a worse disease course, and because inflammation is a key clinical manifestation of severe COVID-19. We will include factors that are known to worsen the prognosis of COVID-19 such as presence of hypertension, obesity, diabetes, chronic kidney disease, and cardiovascular disease.
We have already identified 19 genetic variants that are associated with severe COVID-19 in a discovery study. But to be confident on these results we must redo the analysis in a different population. And this is what we plan to do with the UK Biobank data.
The COVID-19 pandemic has already affected more than 270 million people and more than 5 million have died. While the vaccination has reduced the mortality due to COVID-19, severe cases continue to occur.
Our aim is to validate some of the genetic variants we identified in the discovery study to be associated with COVID-19 severity. Then, we will develop a calculation to predict the expected COVID-19 severity of the patients based on their genetic variants and risk factors. We plan to fulfill these objectives in 6 months.
The results from this project will be validated genetic variants as well as novel variants associated with COVID-19. Another output will be a calculation to obtain the severity risk of COVID-19 patients based on genetic variants and risk factors. With the knowledge from this project it will be possible to design a genetic test to know in advance the severity risk of COVID-19 patients. This information will allow a more personalized treatment of these patients. In addition, the identification of validated and novel genetic variants will open new research lines to design drugs to treat severe COVID-19 and to reuse drugs currently employed to treat other diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cardiovascular-kidney-metabolic-ckm-conditions-sepsis-copd-obesity-and-their-impact-on-life-expectancy-exploring-risk-factors-prognostic-value-and-multimorbidity-effects

Cardiovascular-Kidney-Metabolic (CKM) Conditions, Sepsis, COPD, Obesity, and Their Impact on Life Expectancy: Exploring Risk Factors, Prognostic Value, and Multimorbidity Effects

Last updated:
ID:
617672
Start date:
30 April 2025
Project status:
Current
Principal investigator:
Dr Zhenyu Peng
Lead institution:
Second Xiangya Hospital of Central South University, China

This study investigates risk factors for cardiovascular-kidney-metabolic (CKM) conditions and explores how multimorbidity, including sepsis, chronic obstructive pulmonary disease (COPD), and obesity, impacts life expectancy and mortality. The research seeks to address the following questions: What are the primary risk factors (e.g., adipose tissue distribution, sedentary lifestyle, physical activity levels) for CKM conditions, sepsis, COPD, and obesity? How does the concurrence of CKM diseases with sepsis, COPD, and obesity affect life expectancy and mortality? What roles do modifiable lifestyle factors and body composition play in the progression of these conditions?
Objectives: To identify and quantify modifiable lifestyle risk factors for CKM conditions, sepsis, COPD, and obesity. To examine the impact of concurrent CKM diseases, sepsis, COPD, and obesity on mortality and life expectancy.
To investigate the relationship between body composition (e.g., adiposity) and CKM conditions, sepsis, COPD, and obesity, focusing on physical activity and sedentary behavior.
Scientific Rationale: CKM conditions, sepsis, COPD, and obesity are major contributors to global mortality, significantly reducing life expectancy. With an aging population, multimorbidity involving these conditions poses a critical public health challenge. While traditional risk factors like hypertension and dyslipidemia are well documented, the impact of modifiable lifestyle factors and body composition on these diseases remains underexplored. This study aims to fill these knowledge gaps, providing valuable insights for the prevention and management of CKM multimorbidity and its associated health outcomes.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cardiovascular-kidney-metabolic-health-and-all-cause-and-cause-specific-mortality-in-the-uk-biobank-cohort

Cardiovascular-kidney-metabolic health and all-cause and cause-specific mortality in the UK Biobank cohort

Last updated:
ID:
300908
Start date:
27 November 2024
Project status:
Current
Principal investigator:
Dr Zenghui Zhang
Lead institution:
First Affiliated Hospital of Jinan University, China

There is a high burden of poor cardiovascular-kidney metabolic health in general population, which affects nearly all organ systems and has potent impact on the incidence of cardiovascular disease. More guidance is needed on definitions, staging, prediction strategies, and algorithms for the prevention and treatment of cardiovascular-kidney-metabolic (CKM) syndrome to optimize CKM health across diverse clinical and community settings. Regrettably, there have been limited efforts to translate individual cardiovascular and kidney risk profiles into a visual tool and explore their prognostic roles in long-term outcomes among the general population. In our study, we will use data from UK Biobank cohort to establish a scoring system for CKM consisting of a series of risk factors. Achieving optimal CKM control were associated with lower risk of mortality and longer life expectancy.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cardiovascular-kidney-metabolic-syndrome-and-geriatric-syndrome-multidimensional-analyses-of-health-risks-in-the-middle-aged-and-elderly-population

Cardiovascular-kidney-metabolic Syndrome and Geriatric Syndrome: Multidimensional Analyses of Health Risks in the Middle-Aged and Elderly Population

Last updated:
ID:
292953
Start date:
9 June 2025
Project status:
Current
Principal investigator:
Dr Pan Huang
Lead institution:
Wenzhou Medical University, China

Cardiovascular-kidney-metabolic (CKM) syndrome is a health disorder due to connections among heart disease, kidney disease, diabetes, and obesity, increasing the risk of development and progression of cardiovascular disease and leading to poor health outcomes. Geriatric syndromes!GS! are common conditions that affect older adults and can have a significant impact on quality of life, such as frailty, falls, cognitive impairment, and depression.
People with CKM syndrome are often more likely to develop GS!and GS also promotes the progression of CKM syndrome. The aim of this research project is to investigate the interactions and effects of GS and CKM syndrome on each other. In addition, we will also investigate the risk and protective factors associated with both syndromes, including demographic, lifestyle, health, and genetic factors, and explore the underlying mechanisms that drive their progression over time.
The project is expected to last three to four years, and the findings will have an important impact on public health. By finding the interaction between GS and CKM syndrome and identifying the factors that contribute to the development and progression of GS or CKM syndrome, this study will inform the development of targeted prevention and intervention strategies to improve health status and quality of life in middle-aged and older adults.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cardiovascular-medication-use-for-the-primary-and-secondary-prevention-of-cardiovascular-disease-in-the-uk-trends-determinants-and-lifestyle-behaviours

Cardiovascular medication use for the primary and secondary prevention of cardiovascular disease in the UK: trends, determinants and lifestyle behaviours.

Last updated:
ID:
41437
Start date:
3 September 2018
Project status:
Closed
Principal investigator:
Ms Inna Nicole Thalmann
Lead institution:
University of Oxford, Great Britain

AIMS
The aims of this DPhil research are threefold. First, the study aims to analyse the situation in the UK by estimating the degree of suboptimal cardiovascular medication use for the primary and secondary prevention of cardiovascular disease (CVD) and to examine variations in the uptake of interventions, such as cholesterol- and blood-pressure-lowering drugs, across socio-economic groups.

Secondly, the DPhil study aims to investigate the role of patient characteristics in the underutilisation of cardiovascular medications by studying their relevance to the use of cardioprotective medications among patients with (a) a history of CVD-related events, procedures and operations and (b) individuals at high risk of CVD. The study also aims to examine associations between non-adherence to cardiovascular medications for the secondary prevention of CVD and subsequent adverse cardiovascular events.

Third, the DPhil study aims to investigate lifestyle patterns and clustering of lifestyle behaviours of individuals with a history of CVD or at high risk of CVD, who use cardiovascular medications for the primary or secondary prevention of CVD, compared to those who do not.

SCIENTIFIC RATIONALE
Although there is substantial evidence on the cost-effectiveness of cardiovascular medications such as cholesterol- and blood pressure-lowering drugs in reducing risks of cardiovascular disease (CVD), preventive drug use and compliance rates remain largely suboptimal in the UK and Europe. However, knowledge regarding the underlying reasons for the suboptimal drug use and variations in the uptake of interventions across socio-economic groups, as well as adherence to clinical guidance on lifestyle modifications during medication use are limited.

Building on the gaps in literature, this DPhil thesis aims to use UK Biobank data to provide new insights into important patient characteristics that are needed to understand the underlying reasons for the suboptimal cardiovascular medication use and variations across socio-economic groups, as well as behavioural lifestyle patterns of medication users.

PROJECT DURATION
The project will take place from April 2018 until April 2021, i.e. 36 months. Access to data will be required for an additional 24 months after completion of the project.

PUBLIC HEALTH IMPACT
Addressing the present challenges of CVD prevention is essential to improving the management of disease and to decreasing the high burden and associated costs of CVD, which is a leading cause of mortality worldwide. The findings will support novel effective policies for treatment amelioration and lifestyle management that are urgently needed given the high burden and costs of cardiovascular disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cardiovascular-metabolic-characteristics-of-regular-exercisers-and-the-effect-of-exercise-on-atrial-fibrillationa-cohort-study

Cardiovascular metabolic characteristics of regular exercisers and the effect of exercise on Atrial Fibrillation!A cohort study

Last updated:
ID:
144894
Start date:
12 December 2023
Project status:
Current
Principal investigator:
Professor Fang Wang
Lead institution:
Beijing Hospital, China

Atrial fibrillation (AF) is a common arrhythmia with the incidence increasing multiply with age. Physical activity has a variety of beneficial cardiovascular effects, such as lowering blood pressure levels, improving hyperlipidemia, and increasing insulin sensitivity. Studies showed that the incidence of AF was higher in patients who did not participate in physical activity than in those who did. The odds of progression of atrial fibrillation increased in patients who lacked physical activity. Metabolism is generally an overall term for the organized series of chemical reactions that take place in an organism for the purpose of sustaining life, which includes anabolism and catabolism, the major processes are glucose metabolism, lipid metabolism, and amino acid metabolism. Myocardial metabolism is essential for the pathophysiology of atrial fibrillation, metabolic disorders directly affect the formation of atrial fibrillation substrate. Therefore, the development and progression of AF affect human metabolism and mitochondrial function. In our study, for all included continuous variables, we will analyze after excluding outliers, which are defined as values that are three standard deviations (SD) above or below the mean value, to eliminate the impact of extreme values. Furthermore, the Shapiro-Wilk tests will be applied to check the normal distribution, and for all non-normally distributed continuous variables, the Box-Cox transformation method will be used to transform it into a normal distribution via ‘car’ package in R Studio, and all data are standardized using the z-score method. Continuous variables will be described as means and SDs, whereas categorical variables will be reported as numbers and percentages. Baseline demographic information, physical activity data, laboratory data, imaging data, and atrial fibrillation data will be tested for intergroup differences using chi-square analysis for classified variables or one-way analysis of variance analysis (ANOVA) for continuous variables. We will further expl


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cardiovascular-mortality-according-to-ldl-c-levels-in-lean-individuals-at-low-cardiovascular-risk

Cardiovascular mortality according to LDL-C levels in lean individuals at low cardiovascular risk.

Last updated:
ID:
239827
Start date:
27 May 2025
Project status:
Current
Principal investigator:
Dr Adrian Soto-Mota
Lead institution:
Monterrey Institute of Technology and Higher Education, Mexico

Our research project aims to investigate the impact of low-carbohydrate diets on cardiovascular health, focusing specifically on the relationship between high levels of low-density lipoprotein (LDL) cholesterol and the risk of cardiovascular events in the absence of other common risk factors. Despite the rising popularity of low-carbohydrate diets for various clinical purposes beyond mere weight loss, there is still limited understanding of their long-term effects on cardiovascular health, particularly when it comes to individuals with isolated high LDL cholesterol levels.

The scientific rationale behind our study stems from the observation that while many studies have highlighted the potential cardiovascular risks associated with high LDL levels, few have examined these risks in individuals following low-carbohydrate diets without considering other confounding cardiovascular risk factors. Our preliminary research, involving a cohort of 548 individuals consuming less than 130 grams of carbohydrates per day, indicated a potential inverse relationship between body mass index (BMI) and changes in LDL cholesterol, prompting further investigation.

The project is set to analyse lipid profiles, dietary patterns, and cardiovascular outcomes among our study participants. We aim to use multivariable survival models, to isolate the effects of LDL cholesterol from other potential risk factors, adjusting for variables like biological sex, age, and serum triglyceride levels to ensure the accuracy of our findings.

Ultimately, our research seeks to clarify whether individuals with high LDL cholesterol but no other cardiovascular risk factors face an increased risk of cardiovascular events when adhering to a low-carbohydrate diet. By providing a more nuanced understanding of these relationships, our findings could have significant public health implications, offering valuable insights for healthcare providers and patients alike regarding the safety and efficacy of low-carbohydrate diets for individuals with specific lipid profiles. This could potentially lead to more personalized dietary recommendations, improving cardiovascular outcomes and overall quality of life for those at risk.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cardiovascular-risk-and-risk-prediction-across-all-stages-of-chronic-kidney-disease-in-the-uk-biobank-cohort

Cardiovascular risk and risk prediction across all stages of chronic kidney disease in the UK Biobank cohort

Last updated:
ID:
71725
Start date:
18 October 2021
Project status:
Closed
Principal investigator:
Dr Nynke Halbesma
Lead institution:
University of Edinburgh, Great Britain

Worldwide, around one in ten of the adult population has kidneys that do not work properly and are at high risk of having a heart attack or stroke. Most research has focused on preventing their kidney disease getting worse. Earlier research has shown that these patients do not always get the best treatment to lower their risk of getting heart disease or stroke. The aim of this research, which is expected to be finished within two and a half years, is to find out which patients are at particularly high risk of developing heart disease or stroke using information from the UK Biobank. The results can be used to make risk scores that doctors can use to find patients at high risk of heart disease or stroke and to make sure that they receive the best treatments to prevent these diseases. The ultimate goal is to improve survival and quality of life for people with kidney disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cardiovascular-risk-factors-drug-treatments-and-their-effects-on-alzheimers-disease-and-related-dementias-in-the-united-kingdom-biobank

Cardiovascular Risk Factors, Drug Treatments, and their Effects on Alzheimer’s Disease and Related Dementias in the United Kingdom Biobank

Last updated:
ID:
67926
Start date:
26 April 2021
Project status:
Current
Principal investigator:
Mr Neal Jawadekar
Lead institution:
Columbia University, United States of America

The primary goals of this project are to better understand the effect that cardiovascular risk factors and corresponding treatments have on risk of Alzheimer’s Disease and dementia in the UK Biobank cohort. The literature to date has been inconsistent. While randomized control trials have shown little to no difference in the prevalence of cognitive impairment between medication-users (e.g. for statins and antihypertensives) and placebo, the relatively short follow-up times (4 to 5 years) are limitations of these RCTs in terms of their ability to detect the long-term effects of these drugs on dementia risk. We would also like to examine the causal association between genetically predicted cardiovascular risk factors (e.g. LDL cholesterol and blood pressure) and dementia, as well as how trends in drug prescribing practices have changed over time, in relation to established guidelines by the National Institute for Health and Care Excellence (NICE), when applicable.

Using UK Biobank data, we will study the effect of medication use and cardiovascular risk factors on dementia risk by using two quasi-experimental methods. First, we will utilize regression discontinuity design to assess the unbiased average treatment effect between various treatments and dementia. Second, we will use Mendelian Randomization, a technique which can harness random gene variation to examine the causal effect between genetically predicted CVD risk factors and dementia. We expect our project will take up to 36 months to complete. The potential implications of this project are significant, given that statins and antihypertensives are two of the most widely-prescribed drug classes in the world. In addition, dementia is a debilitating disorder which negatively impacts individuals and their families all over the world.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cardiovascular-risk-prediction-tools-in-rheumatic-diseases

Cardiovascular risk prediction tools in Rheumatic Diseases

Last updated:
ID:
67547
Start date:
22 February 2021
Project status:
Closed
Principal investigator:
Dr David Michael Hughes
Lead institution:
University of Liverpool, Great Britain

Cardiovascular diseases (CVD, heart disease, stroke) are the leading cause of premature death and a major cause of disability. Doctors can use statistical models to predict people’s risk of developing CVD and decide whether to start preventative treatment.
Rheumatic diseases are diverse, including rheumatoid arthritis (RA), psoriatic arthritis, ankylosing spondylitis, for example. Inflammation is a key risk factor for CVDs, so these people have much higher risk. For example, people with psoriatic arthritis have 55% higher risk of CVD. The same is true for other rheumatic diseases.
In RA, existing prediction tools have been shown to under-predict CVD risk, meaning people at risk may miss out on treatment. Despite this, risk prediction tools have not been tested for other rheumatic diseases to see how accurate they are.
Our aim to test the accuracy of several existing general population risk prediction tools in people with rheumatic diseases. If these tools can be used for these people, their care will be improved by promoting use in clinical practice. If they are not valid for people with rheumatic diseases, our study will highlight the need for future research.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/case-control-analyses-in-neuropsychiatric-disorders

Case-control analyses in neuropsychiatric disorders

Last updated:
ID:
63653
Start date:
1 March 2021
Project status:
Closed
Principal investigator:
Professor Gael Nicolas
Lead institution:
Université de Rouen Normandie, France

Knowledge about genetic susceptibility factors in neuropsychiatric diseases has evolved quickly over the past years. Nevertheless, it is still incomplete. Lists of genes implicated in those disorders are not exhaustive while the risk level held by each pathogenic genetic variant, as well as their interactions, are not fully understood. Our team focuses on two common and distinct diseases, carrying an important burden for the society, Alzheimer disease and Autism spectrum disorder. Both disorders are extremely different but share a common determinism: they are considered as complex disorders in the majority of cases with a high genetic component.
For both disorders, we built a cohort of patients and performed whole exome sequencing, i.e. the sequencing of all the coding regions of the genome. We aim to find novel genes with an excess of rare deleterious genetic variants in cases, independently for each disease, as compared to controls representative of the general populations from the UK biobank dataset. Thus, by increasing the list of genes associated with those neuropsychiatric disorders, this project can improve the genetic diagnostics capabilities in a clinical setting in the future.
In addition and unlike ASD, AD is an age-related disorder. Hence, the probability of developing the disease at a given age when carrying certain genetic variants is very important for the future of AD medical prevention. We will use the UKbiobank in comparison to other published resources to refine our estimations of the penetrance of rare genetic variants from well-known associated genes (such as ABCA7, SORL1 and TREM2).
Lastly, we plan to integrate these data with common polygenic risk to decipher the roles of common and rare risk factors in the genetic architecture of these disorders..


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-associations-of-circulating-biomarkers-with-cardiovascular-disease

Causal associations of circulating biomarkers with cardiovascular disease

Last updated:
ID:
13721
Start date:
1 October 2015
Project status:
Closed
Principal investigator:
Professor Themistocles Assimes
Lead institution:
Stanford University, United States of America

The overall goal of this project is to study the causal roles of the 36 biomarkers currently being assayed in UK Biobank for development of coronary heart disease, stroke and heart failure. Knowledge about causal relations of these 36 biomarkers with cardiovascular outcomes will give important insights regarding the etiological understanding of these diseases and accelerate development of new prevention strategies, including druggable targets. Hence, the proposed research does meet UK Biobank’s stated purpose via improving the prevention and treatment of heart disease and stroke. First, we will study associations of 36 circulating biomarkers representing different biological systems with incidence of coronary heart disease, stroke and heart failure.

Second, by combing data from the UK Biobank gene analyses with the biomarker data, we will perform genetic studies across the whole human genome for all 36 biomarkers to establish common genetic variation associated with respective biomarker.

Third, we will perform so called Mendelian randomization analyses to study whether the biomarkers are causally related to coronary heart disease, stroke and heart failure.
Full cohort (n=502,650).


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-associations-of-myocardial-infarction-and-oxidative-stress

Causal associations of myocardial infarction and oxidative stress

Last updated:
ID:
217818
Start date:
7 November 2024
Project status:
Current
Principal investigator:
Mr Jianbo Guo
Lead institution:
University of Hong Kong, Hong Kong

Oxidative stress plays a significant role in the pathophysiological process following MI. Myocardial ischemia and hypoxia after MI can lead to an increase in reactive oxygen species (ROS), and the imbalance between its production and the antioxidant system induce sustained oxidative stress, further aggravating myocardial damage and cardiac remodeling, thereby causing post-MI complications and even heart failure. Thus, assessing oxidative stress-related biomarkers could enhance our understanding of cardiac remodeling post-MI and help stratify patients’ risk for post-MI complications. Besides, these biomarkers could serve as novel therapeutic targets for MI intervention. Previous observational studies have shown that antioxidant enzymes such as glutathione peroxidase9 and heme oxygenase-1 (HO-1) in the blood could predict the prognosis of MI. Regarding treatment, various therapeutic approaches aim to regulate oxidative stress-related cell signaling pathways to effectively treat MI. For instance, melatonin reduces post-MI injury by activating the Notch1/Mfn2 pathway, thereby decreasing oxidative stress. Early administration of melatonin after percutaneous coronary intervention significantly reduces infarct size. Other compounds like wogonin, hirudin, and dapsone could alleviate oxidative stress post-MI and preserve normal myocardial function by targeting the NRF2/HO-1 pathway. Stem cell therapies involving overexpression of HO-1 in stem cells have also shown promise in improving stem cells’ tolerance to hypoxia and oxidative stress, thereby enhancing myocardial function in infarcted myocardium. However, numerous antioxidant enzymes and signaling pathways have not been extensively studied experimentally and clinically. This suggests that the currently monitored oxidative stress biomarkers in post-MI treatment are limited, and related therapies are not fully developed, leaving potential biomarkers unexplored. This study aimed to determine if there were causal relationships between MI and oxidative stress though the mendelian randomization (MR) approach.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-associations-of-novel-and-established-cardiovascular-risk-factors-with-type-2-diabetes-kidney-disease-lung-disease-and-cardiovascular-disease-in-uk-biobank

Causal associations of novel and established cardiovascular risk factors with type 2 diabetes, kidney disease, lung disease and cardiovascular disease in UK Biobank

Last updated:
ID:
59977
Start date:
3 June 2020
Project status:
Closed
Principal investigator:
Dr Ping-Hsun Wu
Lead institution:
Kaohsiung Medical University Chung-Ho Memorial Hospital, Taiwan, Province of China

The aim of this project is to identify the factors that accelerate type 2 diabetes, kidney disease, lung disease and cardiovascular disease Such knowledge will give important insights regarding the etiological understanding of these diseases and help to identify new prevention strategies.
The estimated project duration would be three years. In the beginning, we will study associations of circulating and urinary biomarkers and lifestyle factors with type 2 diabetes, kidney disease, lung disease and cardiovascular disease e. Then we will perform genetic studies using the GWAS genetic data for measured biomarkers to establish common genetic variation associated with respective biomarker and lifestyle factors. Finally, we will perform Mendelian randomization analyses to study whether these biomarkers, and other candidate phenotypes are causally related to type 2 diabetes, kidney disease, lung disease and cardiovascular disease and their risk factors.
Chronic disease is the leading cause of death in UK and globally, and places a huge burden on the afflicted individuals, their families and society as a whole. Therefore, new approaches to prevent and treat these diseases are urgently needed.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-associations-of-novel-and-established-risk-factors-with-type-2-diabetes-kidney-disease-lung-disease-and-cardiovascular-disease-in-uk-biobank

Causal associations of novel and established risk factors with type 2 diabetes, kidney disease, lung disease and cardiovascular disease in UK Biobank

Last updated:
ID:
52678
Start date:
31 October 2019
Project status:
Closed
Principal investigator:
Professor Tove Fall
Lead institution:
Uppsala University, Sweden

The aim of this project is to identify the factors that accelerate type 2 diabetes, kidney disease, lung disease and cardiovascular disease. Such knowledge will give important insights regarding the etiological understanding of these diseases and help to identify new prevention strategies.
The estimated project duration would be three years. First, we will study associations of circulating and urinary biomarkers, physical measurements and lifestyle factors with type 2 diabetes, kidney disease, lung disease and cardiovascular disease. Then we will perform genetic studies using the GWAS genetic data for those risk factors to establish common genetic variation associated with respective biomarker, physical measurement and lifestyle factors. Finally, we will perform Mendelian randomization analyses to study whether these exposures and other candidate phenotypes are causally related to type 2 diabetes, kidney disease, lung disease and cardiovascular disease and their risk factors. Chronic disease is the leading cause of death in UK and globally, and places a huge burden on the afflicted individuals, their families and society as a whole. Therefore, new approaches to prevent and treat these diseases are urgently needed.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-contributors-to-clinical-outcomes-of-sepsis-and-septic-shock

Causal contributors to clinical outcomes of sepsis and septic shock

Last updated:
ID:
164911
Start date:
17 June 2025
Project status:
Current
Principal investigator:
Dr Keith R. Walley
Lead institution:
University of British Columbia, Canada

Infections are the most common cause of death worldwide and no significant improvements in the treatment of severe infection have been developed since the discovery of antibiotics almost a century ago. The role that genetics play in severe infections has not previously been widely investigated. Sepsis is an illness where the patient’s tissues and organs are damaged by the body’s own immune response to infection, and is often life-threatening. Sepsis can lead to septic shock when blood pressure drops and fluid replacement does not improve the condition.

The aim of this project is to explore various ways genetics and epidemiological exposures contribute to sepsis and septic shock. Some of the questions we will be exploring include:
– Do cholesterol-related exposures and their regulatory genetic variants affect survival of sepsis and septic shock patients?
– Are there unique types of sepsis patients? Are there unique types of organ-specific sepsis damage? What can we learn from combining exposure data with genetic and clinical data about organ dysfunction in sepsis? Are there exposures that point to treatment of both organ-specific (e.g., kidney, brain) damage due to sepsis and sepsis and septic shock generally?
– What role do these exposures and their regulatory genetic variants have in differentiating classes of patients? What role do they play in differentiating subclasses (classes of organ dysfunction) in sepsis?
– Are cholesterol-specific exposures enriched in most, if not all, sepsis classes and subclasses? Given the abundance of cholesterol-modulating drugs on the market, can we use any of these existing drugs to be repurposed in sepsis or organ-specific damage in sepsis?

Findings of this project could lead to new treatments for sepsis and septic shock and reduce the death toll caused by these illnesses.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-determinants-of-dementia-with-a-vascular-component-dvc-risk

Causal determinants of dementia with a vascular component (DVC-RISK).

Last updated:
ID:
123335
Start date:
6 December 2023
Project status:
Current
Principal investigator:
Dr Emma Louise Anderson
Lead institution:
University College London, Great Britain

Dementia is a leading cause of death, and the number of cases of vascular dementia is likely higher than what is currently estimated because it often occurs alongside other neurodegenerative diseases such as Alzheimer’s disease. While significant progress has been made in studying the genetic drivers of Alzheimer’s disease, research into the causes of vascular dementia has been lacking.

The previous studies on vascular dementia were too small. They did not include a wide range of patients, which made it difficult to find out, making it difficult to determine the important genes and environmental factors that cause the disease. Understanding the role of blood vessel problems in dementia is important because it makes the memory and thinking problems in diseases conditions like Alzheimer’s even worse.

By conducting a genetic study of dementia patients with vascular pathology, this research will help us understand its causes, genetic (i.e., variations in genes that increase the risk) and environmental (i.e., lifestyle choices, health conditions). Identifying genes that increase the risk, and risk factors that can be changed, will allow us to develop ways to prevent and treat the disease. Additionally, by finding ways to identify people who are at risk early on, we can intervene promptly to slow down or prevent the progression of vascular dementia. Ultimately, this research has the potential to make a big difference in public health by reducing the impact of vascular pathology (i.e., issues related to blood vessels) on dementia, and improving the lives of those affected and their families.

This project will overcome these challenges by doing a detailed study using information about our genes and physical characteristics to find new links and develop ways of predicting who is at a higher risk of getting the disease. Additionally, the project aims to study how genes affect the decline of memory and thinking skills, and find factors that can be changed to reduce the risk of developing vascular dementia.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-discovery-of-digital-biomarkers-to-predict-long-term-mental-and-physical-health

Causal discovery of digital biomarkers to predict long-term mental and physical health

Last updated:
ID:
96553
Start date:
21 February 2023
Project status:
Current
Principal investigator:
Mr Max Moebus
Lead institution:
ETH Zurich, Switzerland

We aim to derive new biomarkers that relate to an individual’s physical as well as mental health. We will assess how the derived patterns predict responses to illness (in particular Covid19) in the future and mortality risks. We will leverage our current research on modeling sleep quality and fatigue based on input from wrist-worn wearable sensors, and we will extend our own smaller studies to a population level using the UK Biobank.

As previous work as well as our own research has identified, (physical and mental) health and well-being are highly subjective. One-week activity recordings allow to paint a much more detailed picture of individuals and their routine in unsupervised settings. We aim to derive new insights based on novel feature extraction methods adapted from our previous research. Besides deriving new biomarkers for health, we also aim to advance existing statistical methodology that currently struggles to detect effects on datasets much smaller than the UK Biobank.
We thus hope that the derived biomarkers contribute to a better understanding of highly-subjective aspects of physical and mental health, which we hope will advance personalized health care in the long run. We hope derived biomarkers will also prove useful for other researchers during future projects using the UK Biobank. We further aim to generate new statistical methodology that enables the discovery and analysis of wearable sensor datasets much smaller than the UK Biobank.

We estimate that we will answer our first research question regarding physical health (mortality prediction, hospital admission, time until recovery) within 12 to 18 months. Answering the following research questions will take significantly less time since data processing and feature extraction will be mostly completed. We estimate that answering research questions regarding mental health and Covid19 response will take around 6 months each. To evaluate causal inference methodology on small longitudinal we estimate another 6 to 12 months, depending on whether we find promising areas for improvement. In total, we thus estimate the project to be finished within 3 years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-effect-of-maternal-vitamin-d-levels-and-body-mass-index-on-pregnancy-related-outcomes

Causal effect of maternal vitamin D levels and body mass index on pregnancy-related outcomes

Last updated:
ID:
23938
Start date:
1 January 2017
Project status:
Current
Principal investigator:
Dr Maria (preferred name Carolina) Carolina Borges
Lead institution:
University of Bristol, Great Britain

We aim to improve the understanding on whether low maternal vitamin D levels and high maternal body mass index (BMI) can contribute to developing adverse outcomes during and after pregnancy, such as gestational diabetes, miscarriages, stillbirth, need for caesarean, low birth weight and longer hospital stays after delivery. Investigating the influence of maternal BMI and vitamin D levels on pregnancy-related adverse outcomes fits with the central aim of UK Biobank to improve prevention and treatment of diseases. For this study, we will use data on BMI, vitamin D levels, genetics, and obstetric-related outcomes in all women that have ever being pregnant. We will compare women with different genetic makeups related to vitamin D and BMI using a technique named Mendelian randomisation to assess whether low maternal vitamin D levels or high maternal BMI are likely to increase the risk of developing adverse pregnancy-related events. Data will be requested for females only (n < 273,000)


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-effect-of-poor-oral-health-on-dementia-and-alzheimers-disease-an-instrumental-variable-approach

Causal Effect of Poor Oral Health on Dementia and Alzheimer’s Disease: An Instrumental Variable Approach.

Last updated:
ID:
918963
Start date:
5 August 2025
Project status:
Current
Principal investigator:
Dr Hyun Ja Lim
Lead institution:
Princeton Pharmatech LLC, United States of America

Dementia and Alzheimer’s disease are one of the main causes of death and disability in the elderly, and currently have no known treatments to prevent or stop it.
The aim of our research is to estimate the causal effect of poor oral health on dementia and Alzheimer’s disease. As of now, scientific literature does not provide conclusive evidence supporting a causal relationship between poor oral health and dementia due to significant bias from unmeasured confounders and reverse causality. This prompts the use of an Instrumental Variable (IV) estimation, which enables us to account for unmeasured confounding, and the use of a prospective follow-up study which eliminates the possibility of reverse causality. In our research, we seek to understand whether the relationship between poor oral health and dementia is causal or spurious, and whether the nature of this relationship extends to more specific cases of oral health, including missing teeth, breathing problems, or periodontal disease, and also specific cases of dementia, including Alzheimer’s disease. Our main research objectives are: (1) understand the current scientific knowledge on this topic and compile a detailed lit review. (2) Identify suitable instrumental variables and test IV strength with UK Biobank data. (3) Perform 2sls with a strong IV, get results, and perform sensitivity analysis. The main mode of disseminating our findings will be through a detailed research paper, which we plan to publish in an accredited journal. We may also present our findings at some Alzheimer’s / Oral health conferences.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-effects-of-environmental-lifestyle-and-genetic-factors-on-chronic-diseases

Causal effects of environmental, lifestyle, and genetic factors on chronic diseases.

Last updated:
ID:
106707
Start date:
16 November 2023
Project status:
Current
Principal investigator:
Professor Nan Li
Lead institution:
Peking University Third Hospital, China

Chronic diseases are a major contributor to disease burden worldwide. In recent years, more studies have explored the association of disease with influencing factors through epidemiological methods such as cohort studies and causal inference methods such as mendelian randomization. Early intervention in chronic diseases can effectively improve the prognosis and reduce the burden. So, this project plans to elucidate the causal relationship between chronic diseases (hypertension, stroke, coronary heart disease, diabetes, dementia, Alzheimer’s disease, Parkinson’s disease, schizophrenia, depression disorders, chronic obstructive pulmonary disease, cancer, etc.) and candidate risk factors, including demographic and sociological characteristics (gender, education, etc.), lifestyle (physical activity, dietary, smoking, etc.), biomarkers (lipids and lipoproteins, sex hormones, etc), and genetics (SNPs, etc.). Our team focuses on the application of statistical methods in medical research. Therefore, in this project, we plan to apply a data-driven strategy. This study will use some appropriate statistical methods to help us discover associations or pathways between above listed factors and chronic diseases. The project will involve about 500000 subjects and a duration of 3 years. We hope to provide more convincing evidence for detecting new risk biomarkers, pathways from the biomarker to disease, and prediction of the diseases. The project is expected to provide potential biomarkers, appropriate statistical methods, and predictive models, and ultimately reducing the global burden of chronic diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-effects-of-maternal-vegetarian-diets-during-pregnancy-on-offspring-health

Causal effects of maternal vegetarian diets during pregnancy on offspring health

Last updated:
ID:
80739
Start date:
1 February 2022
Project status:
Closed
Principal investigator:
Mr Peiyuan Huang
Lead institution:
University of Bristol, Great Britain

Vegetarian diets are defined as diets without the intake of meat, fish, or seafood. Vegetarianism is socially and culturally patterned, and nutrient intakes also differ between vegetarians and non-vegetarians. While previous studies have found that vegetarian diets are associated with a range of health outcomes in the general population, evidence on the relationship between maternal vegetarian diets during pregnancy and offspring health remains scarce. This three-year project is part of a broader project that aims to investigate the influences of maternal vegetarian diets during pregnancy on a range of offspring health outcomes from birth to childhood. Using the UK Biobank data, we aim to identify the genetic determinants of vegetarianism and use them as a “proxy” of maternal vegetarianism to better examine its causal effects on offspring health. Given the increasing popularity of vegetarianism and the importance of optimal nutrition during pregnancy for maternal and child health, our findings will help comprehensively evaluate the pros and cons of the increasingly popular vegetarianism and have implications for dietary counselling and guidelines for pregnant women.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-gene-mapping-for-complex-traits-by-leveraging-genomics-and-proteomics-data

Causal gene mapping for complex traits by leveraging genomics and proteomics data

Last updated:
ID:
102158
Start date:
29 June 2023
Project status:
Current
Principal investigator:
Dr Dan Zhou
Lead institution:
Zhejiang University, China

In this project, we aim to find out which genes are connected to certain diseases by using different methods. We also want to see how these genes might affect our health in multiple ways and how they can be influenced by our environment.
We already know about many genetic markers that are related to common diseases (e.g., type 2 diabetes), but we don’t fully understand the mechanisms. Because of the complexity of the human genome, the physical position of a genetic marker may mislead the identification of its related “driver gene” which causes the disease.
Empirical evidence shows that we will have a hard time identifying a driver gene among a number of “passenger genes” if we purely rely on genetic variants as markers, indicating that more comprehensive approaches are needed. In this project, we will integrate information about genes, proteins, metabolites, and traits to find out which genes are potentially causing the disease. Given the hypothesis that people from different genetic backgrounds may share the same driver genes, we are proposing the use of novel approaches with higher resolution to deduct potential driver genes for common diseases.
The findings of this project could help scientists to have a better understanding of diseases and to find new ways to treat them. This could mean better medicines that work for more people and personalized treatments based on a person’s genes. They could also identify new target genes for existing drugs, which could help repurpose them for different conditions. Overall, this project could help us learn a lot about how our genes affect our health and how we can use this knowledge to improve healthcare.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-genetic-inference-for-risk-prediction-and-discovery

Causal genetic inference for risk prediction and discovery

Last updated:
ID:
65528
Start date:
12 January 2021
Project status:
Current
Principal investigator:
Dr Matthew Davis
Lead institution:
Invitae Corporation, United States of America

By measuring the genomes of a large number of people and recording whether or not they have a disease, it is possible to identify genetic variations correlated with disease risk. Because an individual’s genome is set at birth, such information would enable clinicians to identify at-risk individuals long before that disease is likely to occur. They could then prioritize testing and early interventions in the individuals that are most at risk.

However, correlation does not imply causation. With genetic risk prediction, correlations between genetic variations and disease risk often do not apply outside of the narrow population in which they were measured. For example, if genetic associations were measured for blonde hair in a Scandinavian population, genetic variants that actually caused another prevalent trait in that population, such blue eyes, would be strongly associated with the blonde hair trait. This could hurt prediction in populations where the correlation between blonde hair and blue eyes is weaker. Empirically, genetic predictions learned from individuals of European-descent are less accurate when applied to non-Europeans.

By inferring causal relationships, we avoid the limitations of correlational analyses. Causal relationships hold across different groups of individuals, while spurious correlations do not. Here, causal inference requires a statistical model of how ancestry, environmental factors, and genetic variation interact. Building this model, connecting it to existing knowledge about human biology, and rigorously testing it are the major research aims of this project.

Inferring causal relationships is a more significant theoretical and computational challenge than measuring correlations, so our approach relies on recent breakthroughs in machine learning, statistics, and high-performance computing. Our aim is to test our system head-to-head against existing state-of-the-art genetic prediction algorithms and report whether accuracy and robustness are improved with our approach.

We expect the project to last two years. Our project will produce more accurate, robust, and equitable tools for genetic prediction of disease risk. As a basic methodological tool, it will be applicable to research in public health and biomedicine. In the clinic, it will enable doctors to stratify individuals by disease risk and prioritize screening and early interventions for complex diseases such as breast cancer, type II diabetes, and heart disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-impact-of-multiple-inflammatory-pathways-on-cognitive-function-and-risk-of-dementia

Causal impact of multiple inflammatory pathways on cognitive function and risk of dementia

Last updated:
ID:
13017
Start date:
4 July 2015
Project status:
Closed
Principal investigator:
Dr Stefano Masi
Lead institution:
University College London, Great Britain

Observational studies suggest that inflammation increases the risk of dementia and cognitive impairment. However, whether inflammation is cause or consequence of poor cognitive function remains unknown. Many genetic variants produce lifelong differences in the circulating levels of inflammatory markers. These genetic variants can be used to form scores which can inform on the cumulative impact of each inflammatory marker on cognition. In this project, the association of the genetic scores affecting circulating levels of inflammatory markers with cognitive function will be investigated to assess whether inflammation is causally relayed to higher risk of cognitive impairment. This research proposal perfectly fits with the UK Biobank’s stated purpose as it is based on a novel research approach intended to increase understanding of the mechanisms and molecular pathways leading to cognitive impairment/dementia. In our analysis, we will focus on genetic variants which activate pathways which can be targeted by drugs already available in clinical practice. This work will facilitate both prediction of future risk and the design
of novel therapeutic strategies which aim to prevent cognitive decline and eventual dementia in a short time frame. We will assess the possible causal relationship between genetic variants that are known to affect circulating levels of inflammatory markers with measures of cognitive function. Previous research has identified several genetic variants robustly associated with circulating levels of inflammatory markers. These will be used to form genetic risk scores for several inflammatory pathways. The associations of cognitive function with each genetic score will suggest causal associations between lifelong differences in the inflammatory pathway and cognitive function. When sufficient hospital episodes involving dementia will become available, we will repeat analyses with this binary outcome. Due to the large variability of the results at the neurocognitive tests we expect that data from the full cohort will be required for this project


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-inference-and-risk-prediction-in-complex-diseases-a-multi-modal-approach-integrating-genomics-and-phenomics

Causal Inference and Risk Prediction in Complex Diseases: A Multi-Modal Approach Integrating Genomics and Phenomics.

Last updated:
ID:
520983
Start date:
4 February 2025
Project status:
Current
Principal investigator:
Professor Airu Hsieh
Lead institution:
Tamkang University, Taiwan, Province of China

Complex diseases are influenced by a multitude of genetic, environmental, and lifestyle factors, making causal inference and accurate risk prediction challenging. Our research aims to advance our understanding of complex diseases by integrating multi-modal data from genomics, phenomics, drug, and behavior and developing novel methods for causal inference and risk prediction.
1. Enhancing polygenic risk score (PRS) predictions: We will leverage cross-biobank studies to improve PRS predictions by incorporating biomarkers and disease networks. By accounting for complex interactions between genetic variants, molecular phenotypes, and clinical outcomes, we aim to develop more accurate and clinically actionable PRS.
2. Addressing horizontal pleiotropy in Mendelian randomization (MR): We will propose a novel MR framework that accounts for complex correlated horizontal pleiotropy and time-varying effects through disease-disease networks.
3. Phenome-wide association studies (PheWAS) and MR: We will conduct comprehensive PheWAS to identify novel causal associations between exposures and a wide range of traits. To address challenges in selecting appropriate instrumental variables in MR, we will explore various approaches, including data-driven methods and machine-learning techniques.
4. Disease-disease network: By integrating genetic data with phenotypic data, we aim to discover novel disease associations and develop disease networks.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-inference-for-complex-data-structure-with-mendelian-randomization

Causal inference for complex data structure with Mendelian randomization

Last updated:
ID:
116409
Start date:
6 June 2024
Project status:
Current
Principal investigator:
Dr Lin Liu
Lead institution:
Shanghai Jiao Tong University, China

Data-driven biological research has led to numerous breakthroughs in life sciences and medicine, including helping deliver new treatments for complex diseases and deepening our understanding of biological evolution and disease etiologies. Data-driven biological research certainly calls for better statistical methods for analyzing biological datasets, to ensure that the new knowledge drawn from data analyses is not confounded by the potential violation of the assumptions made or the biases in the data collection process. Good methods should also be able to leverage as much information as possible, including information from other data collection centers, other data modalities, and even existing biological knowledge on the biological system under study. The more information available to us, the less likely we make a mistake. However, the premise is that we have good methods to achieve that.

In this project, our aim is to partially fulfill this goal, by developing rigorous methods for drawing causal conclusions from UK Biobank data, using datasets from multiple centers, multiple modalities, and multiple time points. We expect that our new methods can leverage more information than the existing ones, including datasets and abstract information from databases like KEGG, Gene Ontology, etc. Our goal is to ground the statistical methodologies with rigorous statistical theory, in order to offer biologists convincing evidence from statistical analyses with as small statistical errors as possible. In the long run, we believe that results from such rigorous statistical methods should be more trustworthy. Based on such results, it is also more convincing to our “consumers”, such as decision-makers, and physicians, to use the new knowledge when facing important public health questions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-inference-for-disease-effect-on-cancer

Causal inference for disease effect on cancer

Last updated:
ID:
64071
Start date:
26 October 2021
Project status:
Current
Principal investigator:
Dr Rachel Dania Melamed
Lead institution:
University of Massachusetts Lowell, United States of America

Cancer seems to arrive as a sudden calamity, but most cancers develop over a period of many years. Understanding factors that influence cancer development can help us predict or prevent cancer. As well, increasing our knowledge of cancer biology can help us treat it. One such influence on cancer is a history of certain common diseases, such as obesity and diabetes. A number of relationships of cancer with rare and common diseases have been reported, but so far no comprehensive study has sought to systematically identify changes in cancer incidence due to onset of common disease. In this study, we will perform a systematic assessment of the risk of cancer due to other common diseases using health data. Then, we will use genetic data as an independent way to estimate the effect of common diseases on cancer, as it is subject to a different set of biases. Finally, genetic data will provide a basis for identifying the biological processes altered by these diseases that impacts cancer incidence. This project will provide insight into cancer risk for people who suffer other diseases, potentially informing screening guidelines and precision medicine prognoses. As well, the results will illuminate the biology of the early stages of cancer development. The project also has the potential to suggest personalized therapies, as common inherited genetics will be mapped to specific cancer processes.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-inference-in-large-scale-clinical-neuroimaging-studies

Causal Inference in Large-Scale Clinical Neuroimaging Studies.

Last updated:
ID:
36681
Start date:
29 October 2018
Project status:
Current
Principal investigator:
Dr Christopher Long
Lead institution:
Bournemouth University, Great Britain

According to the World Health Organisation (2012), neurological disease in its various forms afflicts tens of millions of people worldwide and in some of its domains is forecast to rise significantly. In prevalence studies of Alzheimers Disease (AD) for example, figures are predicted to rise from around 36 million today to over 100 million by 2050. While there are limited treatment options available for many of these afflictions, it is likely that future treatment strategies will be most effective if applied at the earlier stages of disease.
It is the aim of this one year project to increase the clinical utility of large-scale neuroimaging datasets through improved statistical modelling of underlying disease factors as they relate to neurological disease. With the ultimate goal of developing a statistical framework for assessing different treatment regimens, we seek both to enhance early disease detection and advance understanding of the complex neurological mechanisms that foreshadow disease onset.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-inference-study-to-clarify-the-association-between-alcohol-consumption-diabetes-cancer-and-cardiovascular-disease-paradox

Causal inference study to clarify the association between alcohol consumption, diabetes, cancer and cardiovascular disease paradox

Last updated:
ID:
66486
Start date:
7 April 2021
Project status:
Current
Principal investigator:
Professor Sun Ha Jee
Lead institution:
Yonsei University, Korea (South)

The controversy over the association between alcohol consumption and cardiovascular disease is a researcher’s concern and national concern. The best way to confirm causality is RCT(Randomised control trial). Still, it is challenging to implement as an ethical issue, and the Mendelian Randomization study is drawing attention as a new way to replace it.
Through a large-scale Korean cohort (KCPS-II) collected for about a decade, the study plans to analyze epidemiologically, taking into account known risk factors for cardiovascular diseases, such as drinking, smoking, age, physical activity, blood pressure, cholesterol, and blood sugar.
Of the approximately 150,000 people we have obtained blood samples, and consent forms, a genome-wide association study (GWAS) analysis will be conducted with the data of the subjects from which genetic data has been collected. Through this, we want to discover the genes associated with alcohol consumption.
The KCPS-II is a large-scale Korean cohort that includes blood samples collected from 2004 to 2013 and various clinical and health information. According to the data, not only the prevalence at the time of blood collection but also the new disease and death data that have occurred since then by follow-up observations. It is possible to discover the disease risk factors and biomarkers and construct them a disease prediction model for Koreans.
It is intended to construct a predictive model of cardiovascular disease using the fusion interaction of environmental factors, biomarkers, and genetic factors from epidemiological analysis of long-term collected cohort data.
Today, precise medical techniques tailored to individuals are required, and the need for multidisciplinary research is increasing. The predicted models built through this process will be verified using UK Biobank data targeting the European population.
Through UK biobank, which has large-scale genetic information, we expect cooperation and development on research methods to identify the relevance of disease via big data.
The interaction of various genetic-environment factors determines the occurrence of cardiovascular disease. A wide range of surveys and tests involving various variables are needed, and genetic information is required to discover Korean specific genetic factors.
Based on KCPS-II Biobank data meeting all these conditions, it is expected to play an essential role in evaluating the clinical usefulness of cardiovascular diseases as well as in the mechanism study of disease occurrence and the development of potential therapeutic materials.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-inference-using-genetic-and-genomic-data-to-understand-complex-diseases

Causal inference using genetic and genomic data to understand complex diseases

Last updated:
ID:
221732
Start date:
8 October 2024
Project status:
Current
Principal investigator:
Dr Manikandan Narayanan
Lead institution:
Indian Institute of Technology Madras, India

In our research project, we aim to arrive at a holistic understanding of complex diseases such as diabetes, cardiovascular diseases and neurological disorders. We approach the problem at hand via two different ways. Firstly, we intend to investigate the similarities and differences in the behaviour of mutations (SNPs) across different populations and ethnic groups of the world to come up with population specific diseases risk scores. These risk scores (called PRS) are single valued estimates that, given the genomic information of an individual, can give us an idea about how predisposed they are to develop a certain disease. We aim to refine these PRS to take into the account the differences in the genetic makeup of people across populations and the disparity in the amounts of available data per population per disease. Secondly, we intend to explore the underlying causality behind the occurrence of these complex diseases. Studying and identifying the truly causal factors for a disease is of much more importance than merely identifying associations between them. We aim to employ the recent advancements in the area of causal inference and causal discovery to come up with insights that would be essential to make sense of the data, to guide actions and policies and to learn from the resulting successes or failures in a way that traditional tried-and-tested statistical and machine learning methods fail to do. Taking into account that some preliminary analysis and model development has already been carried out at our end, we estimate that this project would require a duration of 3 years. Throughout this project, we hope to further advancements in predictive, preventive and personalised medicine in underrepresented and understudies populations of the world.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-interactions-in-physical-activity-and-premature-cardiometabolic-mortality

Causal interactions in physical activity and premature cardiometabolic mortality

Last updated:
ID:
53710
Start date:
12 June 2020
Project status:
Current
Principal investigator:
Dr Elina Sillanpää
Lead institution:
University of Jyvaskyla, Finland

Physical activity has been highlighted as a cost-effective strategy for prevention of cardiometabolic diseases. To date policy actions and treatments are based on an assumption of a similar beneficial effect from physical activity on the population. Human disease development is, however, a complex interplay between genetic inheritance, life style and environmental factors. This project investigates how genetics modulate level of physical activity and disease risk. We will also examine if and how physical activity mediates the realization of genetic risk in cardiometabolic diseases. Results can be used in developing targeted and effective lifestyle interventions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-mediation-analysis-with-machine-learning-to-understand-the-increased-risks-of-depression-in-overweight-and-obese-population

Causal mediation analysis with machine learning to understand the increased risks of depression in overweight and obese population

Last updated:
ID:
99946
Start date:
29 March 2023
Project status:
Current
Principal investigator:
Professor Henry Tong
Lead institution:
Macao Polytechnic University, China

Worldwide obesity rates have almost tripled since 1975. At the same time, rates of depression have steadily risen. Previous studies have established a link between these two conditions, showing that the prevalence of depression in people with obesity is twice as high as in people of a healthy weight. In terms of treatment, some medications used to reduce appetite for people with obesity have been implicated in the etiology of depressive symptoms. On the other hand, weight gain is a side effect of almost every antidepressant. You take the medication to reduce depression, but it causes you to gain weight which worsens your depression. The vicious cycle of depression and obesity remains largely unknown.
In this project, we aim at unveiling the causal mechanisms of increased risks of depression in overweight and obese population through causal mediation analysis with machine learning. The specific research questions to be answered include:
(1) Is the marriage status (Single, married, divorced, or widowed) associated with the increased risks of depression in overweight or obese people?
(2) Is there a potential association between the usage of diet pills for weight loss and the increased risks of depression in the population?
(3) What are the biochemical, metabolic, environmental, or genetic factors that mediate the increased risks of depression in overweight or obese people?


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-pathways-in-ischemic-stroke-prognosis-an-integrated-mendelian-randomization-study-of-clinical-genetic-and-modifiable-factors

Causal Pathways in Ischemic Stroke Prognosis: An Integrated Mendelian Randomization Study of Clinical, Genetic, and Modifiable Factors

Last updated:
ID:
1038426
Start date:
17 October 2025
Project status:
Current
Principal investigator:
Mr Jinyu Lin
Lead institution:
The Second Affiliated Hospital of Shantou University Medical College, China

Research Questions:
Do key clinical, lifestyle, and genetic factors have a true !causal relationship! with functional outcomes after ischemic stroke (IS)?
Aims:
Aim 1: To quantify the causal effects of genetically proxied clinical traits (e.g., blood pressure, lipids) and lifestyle factors (e.g., physical activity) on post-stroke outcomes (e.g., 3-month mRS score) using a Mendelian Randomization (MR) framework.
Aim 2: To investigate the interactions between polygenic risk scores (PRS) and modifiable risk factors (e.g., exercise) and their combined effects on prognostic outcomes.
Aim 3: To integrate causal risk factors into a clinico-genetic risk prediction model for stroke prognosis, followed by internal validation.
Objectives:
To identify and validate at least 2-3 modifiable factors (e.g., Lp(a) levels, moderate-to-vigorous physical activity) with significant causal effects on stroke prognosis.
To develop an open-source risk scoring tool to aid clinicians in the early identification of patients at high risk of disability.
To provide prioritised causal evidence for designing future targeted interventional trials for stroke rehabilitation.
Scientific Rationale:
Ischemic stroke constitutes 87% of global stroke burden. Despite 40% of ischemic stroke survivors developing functional disability (mRS!3), observational studies suffer from residual confounding. MR overcomes this by leveraging genetic variants as instrumental variables. The current research still lacks a causal assessment of dynamic lifestyle factors (such as the activity intensity monitored by accelerometers) and ischemic stroke. UK Biobank’s trio of genomic data, accelerometry-based activity metrics, and linked hospital records provides unprecedented resources to investigate causal pathways in stroke prognosis.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-pathways-linking-diet-nutrition-and-cardiometabolic-diseases

Causal pathways linking diet, nutrition and cardiometabolic diseases

Last updated:
ID:
77447
Start date:
26 January 2022
Project status:
Current
Principal investigator:
Dr Christine Delon
Lead institution:
NNEdPro, Great Britain

Cardiovascular diseases (CVD), such as strokes and heart attacks, cause millions of deaths every year across the world. Factors such as high blood pressure, high blood sugar, high body weight and smoking can increase CVD risk. Therefore, treating these risk factors could reduce CVD disease and death. Individual risk factors for CVD often influence each other. For example, obesity is associated with increased blood pressure, reduced response to insulin and disrupted cholesterol, all of which contribute to CVD risk.
Diet can influence several risk factors for CVD, as such, it can be a very cost-effective tool for CVD prevention and treatment. Similar to other factors related to CVD, nutrients consumed as a part of individual’s diets interact among themselves and also with different processes in the body that contribute to CVD risk. For example, consumption of diets that are rich in fibre can have a beneficial impact on blood glucose and cholesterol levels. Understanding which nutrients or particular diets are the cause of changes observed in CVD risk is essential to inform guidelines and practices in CVD prevention and management and therefore have important public health value.
Establishing whether particular diets or nutrients are the cause of CVD would traditionally require researchers to randomly assign individuals to particular types of diets however, this is not always feasible or practical in nutrition research, particularly in the CVD domain. For instance, it can take several years before CVD outcomes can be directly measured. Observing the occurrence of CVD among a group of individuals is an alternative strategy but it also has some limitations. Using new ways to analyse observational information, using tools borrowed from the causal inference thinking, can help to address some of these limitations. Therefore, the overall aim of this project is use tools from causal inference to disentangle the relative importance of particular dietary factors on CVD risk, but also the connections and pathways between nutrients and diets and CVD risk.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-pathways-of-environmental-and-hereditary-risk-in-common-diseases-observational-metabolomic-and-genetic-studies

Causal pathways of environmental and hereditary risk in common diseases: observational, metabolomic, and genetic studies

Last updated:
ID:
104807
Start date:
24 October 2023
Project status:
Current
Principal investigator:
Professor Børge Grønne Nordestgaard
Lead institution:
Copenhagen University Hospital, University of Copenhagen, Denmark

Most deaths world-wide result from common diseases, including heart attack, stroke, cancer, chronic lung disease, and diabetes. These and other diseases are steadily becoming more common, partly because more people reach higher ages, but also because more people world-wide smoke, are exposed to environmental pollution, eat more processed food, and exercise less.
The aims of this study are to:
– Identify new risk factors that cause common diseases.
– Uncover more about the ways in which known risk factors cause common diseases.
– Describe the extent of the harm caused by specific risk factors, in terms of the number of people that become ill due to exposure.
Scientific rationale:
Earlier population studies have led to discovery of risk factors that cause disease, as for example elevated cholesterol that causes heart attack and stroke, genetic mutations that cause cancer, smoking that causes chronic lung diseases, and obesity that causes diabetes. This has spurred interventions for dealing with these risk factors, which has prevented many individuals from developing disease. However, many individuals still develop common diseases, and more work therefore needs to be done to identify new risk factors, learn more about existing risk factors, and describe the potential benefit of intervening to decrease such risk factors.
Risk factors that cause disease can be identified by studying the association between randomly distributed genetic variation associated with such risk factors. If there is an association, it can be tested if there is also an association between the genetic variation and a disease. If there is, it can in some cases be inferred that the risk factor causes the disease. This method is called Mendelian Randomization and is best when done in large population studies like the UK biobank, in which there are many people that develop disease due to different causes.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-pathways-of-lipoproteins-in-atherosclerotic-cardiovascular-diseases-and-cause-specific-mortality-observational-metabolomic-and-genetic-studies

Causal pathways of lipoproteins in atherosclerotic cardiovascular diseases and cause-specific mortality: observational, metabolomic, and genetic studies

Last updated:
ID:
96405
Start date:
7 December 2022
Project status:
Current
Principal investigator:
Dr Benjamin Nilsson Wadström
Lead institution:
Herlev and Gentofte University Hospital, Denmark

Atherosclerotic disease, leading to clogged blood vessels in the heart, brain, and legs, is the main cause of mortality world-wide. Atherosclerotic disease is becoming more common, partly because more people reach higher ages, but also because more people smoke, are exposed to environmental pollution, eat more processed food, and exercise less. Elevated low-density lipoproteins carry cholesterol in the bloodstream and can accumulate in the blood vessel walls, thereby causing atherosclerotic disease. There are also other lipoproteins, called remnant lipoproteins, that cause atherosclerosis and increase inflammation in the body. However, the mechanism behind this is not known, nor is it known if different lipoproteins affect blood vessels in the heart, brain, and legs the same. Furthermore, it is not known if individuals with elevated remnant lipoproteins are at higher risk of dying from causes not related to atherosclerosis.

Aims:
The aims of this study are to:
– Determine if different lipoproteins may similarly cause atherosclerotic disease in the heart, brain, and legs.
– Determine if different lipoproteins may cause cardiovascular, cancer and other mortality.
– Determine if different lipoproteins may similarly cause atherosclerotic disease in the heart, brain, and legs and cardiovascular, cancer and other mortality in individuals with diabetes.

Scientific rationale:
Drugs that can lower remnant lipoproteins, in addition to low-density lipoproteins, are likely to soon become clinically available. This opens the possibility to identify groups that may especially benefit from lowering of remnant lipoproteins in relation to low-density lipoproteins.
This can be done using a method called Mendelian Randomization, which and can be used to determine causality of risk factors such as low-density lipoproteins and remnant lipoproteins. It is best when done in large population studies like the UK biobank, in which is possible to compare the people that develop different atherosclerotic diseases and die from different causes.

Project duration:
For a start 36 months, may thereafter be extended.

Public health impact:
We expect to provide knowledge for improved prevention of atherosclerotic diseases and death from different causes. Our results may help scientists develop new preventive treatments, clinicians identify which lipoprotein to treat, individuals change lifestyle, and policymakers design public health interventions. Ultimately, we hope our research will contribute to fewer people suffering from disease and more people living longer healthier lives.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-relationship-between-environmental-risk-factors-and-cognitive-impairment-and-underlying-mechanisms

Causal Relationship between Environmental Risk Factors and Cognitive Impairment and underlying mechanisms

Last updated:
ID:
734491
Start date:
12 May 2025
Project status:
Current
Principal investigator:
Mr Enmin Ding
Lead institution:
Chinese Academy of Medical Sciences &Peking Union Medical College, China

Cognitive impairment such as Alzheimer’s disease pose major global health threats, with continuously increasing incidence and mortality rates. Although previous studies have disclosed the association between environmental factors and cognitive impairment, determining the causal relationships is challenging due to issues such as confounding factors and reverse causation.
This study intends to employ Mendelian Randomization to explore whether there exist causal relationships between environmental risk factors (such as air pollution, noise exposure, non-optimal temperature, and social events) and the occurrence and development of cognitive impairment and underlying mechanisms. Firstly, we will identify genetic variants associated with environmental factors from relevant genome-wide association studies. Then, we will make use of a vast amount of information on participants, including environmental exposures, questionnaire information, health records, and multi-omics data to better understand the underlying mechanisms. If the genetic variants are associated with both environmental exposure and cognitive impairment, then we can conclude that environmental exposure is a causal risk factor for cognitive impairment.
The primary objective of this study is to clarify the causal relationships between environmental risk factors and the risk of cognitive impairment. Understanding these causal relationships will provide a scientific basis for the development of targeted prevention and intervention strategies.
We propose to complete this project in 24 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-relationship-between-epicardial-adipose-tissue-and-major-adverse-cardiovascular-events

Causal relationship between epicardial adipose tissue and major adverse cardiovascular events

Last updated:
ID:
144873
Start date:
4 April 2024
Project status:
Current
Principal investigator:
Dr Ming Liu
Lead institution:
Jiangsu Province Hospital of Chinese Medicine, China

Epicardial adipose tissue (EAT), referring to the visceral fat deposit between the outer wall of the myocardium and the visceral layer of the pericardium, was clinically measured by echocardiography, computed tomography volumetric quantification and magnetic resonance imaging(MRI). More and more evidence showed that EAT is a useful and significant imaging biomarker for predicting the risk of a variety of major adverse cardiovascular events (MACE), which was defined as cardiac death, myocardial infarction (MI), unstable angina, coronary revascularization, ischemic stroke, or heart failure. However, the observational studies used to date have limitations that restrict their ability to establish causality, such as unmeasured confounding.

This research project aims to investigate the causal relationship between increased EAT thickness or volume and higher risks of MACE. We will conduct a Mendelian Randomization study by using genetic variant or other mediator which is associated with both EAT thickness and MACE occurrence as an instrumental variable, while controlling for potential confounding factors in UK population..

This study is the first to use the UK Biobank to investigate the effects of EAT thickness or volume on the incidence rate of MACE, providing a novel and unconventional perspective and more comprehensive understanding on the pathophysiology of major cardiovascular diseases. Such program could help us understand if EAT is causally linked to MACE morbidity, which will inform the development of novel intervention strategies including screening for early detection and therapeutics to reduce the burden of cardiovascular disease in UK population. Our program proposes to be finished within 2-3 years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-relationship-between-psychosocial-factors-and-cancer-prognosis-mediated-by-gene-expressions

Causal relationship between psychosocial factors and cancer prognosis mediated by gene expressions

Last updated:
ID:
96744
Start date:
4 April 2023
Project status:
Current
Principal investigator:
Jinfeng Xu
Lead institution:
City University of Hong Kong, Hong Kong

It was previously shown that psychosocial factors affect expressions of some genes, and expressions of some other genes affect the prognosis of cancer patients. However, it remains unclear whether psychosocial factors are independent risk factors to cancer prognosis. To understand the role of psychosocial factors in cancer prognosis, we propose to (i) investigate the prognostic factors of cancer patients, including sociodemographic characteristics, lifestyle and environmental exposures, psychosocial factors and gene expressions; (ii) decipher the direct effects of psychosocial factors and their indirect effects through gene expressions on cancer prognosis by adjusting for potential confounders. The proposed study will contribute towards UK Biobank goals of improving public health in several ways. First, this work will provide new insight of the prognostic factors of cancer patients, which would have implications on multiple tumors. Second, our findings will contribute to informed interventions aimed at susceptible populations with psychosocial factors and will establish a risk prediction model for target population for cancer early detection. The proposed study will utilize the full cohort with available baseline characteristics, sociodemographic characteristics, lifestyle and environmental exposures, psychosocial factors, genome-wide gene expression data and cancer outcomes.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-relationship-between-reduced-kidney-function-and-cancer-risk-a-mendelian-randomisation-study

Causal relationship between reduced kidney function and cancer risk: A Mendelian Randomisation study

Last updated:
ID:
78949
Start date:
24 February 2022
Project status:
Current
Principal investigator:
Miss Ellen Louise Kate Dobrijevic
Lead institution:
University of Sydney, Australia

Chronic kidney disease (CKD) is a major global health problem, involving reduced kidney function and affecting around 13% of the population. CKD is increasingly being identified as a risk factor for cancer and cancer death. Multiple mechanisms have been proposed to explain this observed association. However, the observational studies used to date have limitations that restrict their ability to establish causality, such as unmeasured confounding and reverse causation. This study will use Mendelian Randomisation to examine whether there is a causal relationship between reduced kidney function and the occurrence of cancer and cancer-related mortality.

We will use two data sources to conduct a Mendelian Randomisation study. We will identify genetic variants associated with CKD from the CKDGen consortium published meta-analysis of genome wide association studies. We then propose to use the UK Biobank dataset to investigate if these genetic variants are associated with an increased risk of cancer. If the genetic variants are associated with both CKD and cancer, then we can conclude that CKD is a causal risk factor for cancer. This is a valid conclusion given the design of the study, because genetic variants are randomly allocated at birth and not influenced by behavioural, socioeconomic or physiological factors.

This study aims to investigate the causal relationship between reduced kidney function and all-cause and site-specific cancer risk and death. Understanding if CKD is causally linked to cancer will inform the development of novel intervention strategies including screening for early detection and therapeutics to reduce the burden of cancer in the at-risk population.

We propose to complete this project in 24 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-relationships-between-different-exposures-and-major-metabolic-and-cardiovascular-diseases

Causal relationships between different exposures and major metabolic and cardiovascular diseases

Last updated:
ID:
68376
Start date:
26 January 2021
Project status:
Current
Principal investigator:
Professor Huijie Zhang
Lead institution:
Southern Medical University, China

Noncommunicable diseases including metabolic and cardiovascular diseases, including obesity, diabetes, non-alcoholic fatty liver disease (NAFLD), metabolic syndrome, and coronary heart disease (CHD), account for more than 70% of the deaths worldwide. A variety of exposures, including dietary habits, exercise behavior, smoking, alcohol consumption, nutrient intake, beverage intake and sleep are related with the risk of these diseases. Thus, it is imperative to develop better strategies to reduce the morbidity and mortality of major metabolic and cardiovascular diseases by determining the causal associations between different exposures and major outcomes.
We aim to explore the associations between different exposures and metabolic and cardiovascular disease in a large population-based cohort study. Our project will provide evidence for developing efficient strategies to reduce morbidity and mortality of metabolic and cardiovascular diseases. This project is expected to last for 36 months. The findings may deepen the understanding of associations between different exposures and metabolic and cardiovascular diseases and their related outcomes, and may provide strong evidence for the prevention and therapeutic strategy of metabolic and cardiovascular diseases, and even make a significant contribution to global public health.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-relationships-between-environmental-exposures-and-risk-of-human-cancers-a-phenome-wide-mendelian-randomization-analysis

Causal relationships between environmental exposures and risk of human cancers: a phenome-wide mendelian randomization analysis

Last updated:
ID:
45973
Start date:
20 December 2018
Project status:
Closed
Principal investigator:
Professor Ben Zhang
Lead institution:
Sichuan University, China

Cancer is a leading cause of human death worldwide. Environmental factors play a major role in the development of most types of human cancers. Over the past century, epidemiological studies particularly large prospective cohort studies have linked many environmental factors with risk of human cancers. Observational studies can identify epidemiological associations between environmental exposures and risk of diseases such as cancer. However, whether these exposures directly cause cancer or are simply a risk factor of cancer are largely unclear perhaps due to potential confounders or reverse causality. Thus far, only a few environmental factors have been found to be causally associated with risk of specific cancers, for example, cigarette smoking and risk of lung cancer. Because a lot of environmental factors are modifiable, prevention or early intervention of them may eventually reduce cancer incidence if the associations between these factors and cancer are causal. In this project, we aim to systematically evaluate the potential causal relationships between well-established environmental factors and human cancers. To address this issue, will carry out a prospective cohort study, a phenome-wide association study, and a mendelian randomization analysis using data from the UK Biobank, our group, other public available datasets and published studies. The potential causal effect of a specific exposure (risk factor) on the risk of over cancer or an individual cancer (outcome) is estimated as the ratio of the coefficient of the association between genetics and outcome to that of the association between genetics and the risk factor. We also investigate associations of cancer susceptibility variants with environmental exposures to test for potential reverse causality. We will start analyses as soon as data are available and plan to finish this project and send manuscripts to authors for review within 36 months after we receive the data. We hope that this study will provide a profile of causal relationships between environmental exposures and cancer risk and may help identify novel biological pathways and therapeutic targets for improving prevention and treatment of cancer.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-relationships-between-infectious-and-non-communicable-common-disease

Causal relationships between infectious and non-communicable/ common disease.

Last updated:
ID:
22881
Start date:
1 October 2016
Project status:
Current
Principal investigator:
Professor Chirag Patel
Lead institution:
Harvard Medical School, United States of America

Many non-communicable diseases (e.g., Type 2 diabetes [T2D] and heart disease ) are the most burdensome diseases in the world and are caused by the interplay between environmental exposures and inherited genetic factors. An omni-present exposure includes bacterial agents, such as Helicobacter Pylori (H.Pylori), and viral agents, such as human immunodeficiency virus (HIV). In this investigation, we aim to test whether genetic susceptibility to infection is associated with disease, such as T2D and heart disease (including stroke), and risk factors for these diseases such as body mass index (BMI), cholesterol, and inflammatory biomarkers. Our research is focused on deciphering the risk factors for type 2 diabetes, and heart disease. This focus is in-line with the UK Biobank’s vision of improving the prevention, diagnosis and treatment of a wide range of serious illnesses which include diabetes and heart disease. We will associate genetic variants for susceptibility of infection with common diseases including time to type 2 diabetes. We will also associate variants with body mass index, blood pressure, and other biomarker risk factors for diabetes such as hemoglobin A1C, cholesterol. First we will collect variants that have been previously associated with infectious disease in the GWAS catalog. Second, we will test, using GWAS arrays on the UK Biobank participants, associations between infectious disease SNPs with the time-to-disease. Full cohort.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-relationships-between-lifestyle-and-environment-exposures-and-neurodegenerative-and-cerebrovascular-diseases

Causal relationships between lifestyle and environment exposures and neurodegenerative and cerebrovascular diseases

Last updated:
ID:
94166
Start date:
21 November 2022
Project status:
Current
Principal investigator:
Mr Fabin Lin
Lead institution:
Fujian Medical University, China

Neurodegenerative and cerebrovascular diseases, including Parkinson’s, Alzheimer’s, amyotrophic lateral sclerosis, frontotemporal dementia, vascular dementia, multiple system atrophy, progressive supranuclear palsy, sleep behavior disorder, secondary Parkinson’s disease, cerebral infarction, etc., its economic burden accounts for more than 50% of the world’s medical expenditure.
Lifestyle and environment exposures, including dietary habits, exercise behavior, smoking, alcohol consumption, nutrient intake, beverage intake, and sleep have been associated with the risk of these diseases. We aim to explore the associations between Lifestyle and environment exposures and neurodegenerative and cerebrovascular diseases in a large population-based cohort study. Our project will provide evidence for developing efficient strategies to reduce morbidity and mortality of neurodegenerative and cerebrovascular diseases. This project is expected to last for 36 months.
The findings may deepen the understanding of associations between different exposures and neurodegenerative and cerebrovascular diseases and their related outcomes, and may provide strong evidence for the prevention and therapeutic strategy of neurodegenerative and cerebrovascular diseases, and even make a significant contribution to global public health.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-research-on-genetic-factors-and-lifestyle-habits-in-relation-to-various-chronic-diseases

Causal research on genetic factors and lifestyle habits in relation to various chronic diseases

Last updated:
ID:
117401
Start date:
4 April 2024
Project status:
Current
Principal investigator:
Dr Jie Gao
Lead institution:
The First Affiliated Hospital, Guangzhou University of Chinese Medicine, China

There is a universal causal relationship between lifestyle habits such as diet, physical activity, and social engagement, and several globally prevalent chronic diseases including dementia, cardiovascular and cerebrovascular diseases, diabetes, kidney diseases, gynecological diseases and cancers. By understanding these causal connections and assessing their risks comprehensively, we can gather evidence for the prevention and treatment of chronic diseases. However, it is important to note that these diseases are also closely associated with genetic factors. The complexity of studying causal relationships is further compounded by confounding factors in cross-sectional or retrospective cohort studies. To overcome this challenge, we propose employing Mendelian randomization methods to establish causal relationships specifically between various lifestyle habits and chronic diseases.
This project aims to explore the causal effects of lifestyle factors (such as diet, behavior patterns, and natural environment) and health attributes (including psychological conditions, blood markers, physical measurements, and medical history) on chronic diseases. By incorporating genetic factors and utilizing Mendelian randomization models, we will evaluate the causal effects, mediation effects, and dose-response relationships through various models, including linear models and composite nonlinear models. The ultimate goal is to enhance the understanding of the causal relationship between lifestyle habits and chronic diseases. The conclusions drawn from this research will help healthcare professionals improve chronic diseases by adjusting daily lifestyle behaviors. The duration of this project will be 36 months, and we plan to utilize the entire cohort sample.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causal-studies-of-multiomics-genetics-lifestyle-and-environmental-factors-on-the-progression-and-mortality-of-major-common-metabolic-diseases

Causal studies of multiomics, genetics, lifestyle, and environmental factors on the progression and mortality of major common metabolic diseases

Last updated:
ID:
262612
Start date:
10 October 2024
Project status:
Current
Principal investigator:
Dr Zongji Zheng
Lead institution:
Nanfang Hospital, Southern Medical University, China

Metabolic diseases pose a serious obstacle to the development of global public health, in which lifestyle exposure and multiomic imbalance play a crucial role. Therefore, it is of great significance to integrate the study of common exposures and multi-omics data analysis for the prevention and treatment of metabolic diseases. Multiple guidelines such as KDIGO also advocate lifestyle modifications to reduce the risk of metabolic diseases, while multi-omics techniques can help to fully understand the genetic and metabolite mechanisms behind high-risk life exposures, which can provide patients with more effective life interventions.
Based on the above situation, this study aims to link exposures such as diet, activity and sleep with the metabolic risk of multi-omics imbalance with the help of high-quality data from the UK Biobank, and deeply reveal the intrinsic relationship between life exposure factors and multi-omics imbalance through omics research methods, so as to formulate personalized prevention strategies for patients.
This study will use a linear mixed model to explore the interactions between omics, including life exposures, and their impact on the risk of metabolic disease morbidity. This research will help to understand the causes of metabolic diseases, identify more biomarkers, and inform individualized and highly effective life interventions, ultimately reducing the global public health burden.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causes-and-consequences-of-digestive-diseases-a-prospective-cohort-study-and-a-phenome-wide-mendelian-randomization-analysis

Causes and consequences of digestive diseases: a prospective cohort study and a phenome-wide mendelian randomization analysis

Last updated:
ID:
47515
Start date:
14 February 2019
Project status:
Closed
Principal investigator:
Professor Ben Zhang
Lead institution:
Sichuan University, China

Digestive diseases (ICD-10, codes K00-K93) are a leading cause of morbidity, hospital admission and economic burden worldwide. The pathogenesis of these diseases is understudied. Epidemiological studies have linked a number of environmental exposures with risk of different digestive diseases. Meanwhile, studies have also found that certain digestive diseases may be risk factors for other complex diseases. In addition, digestive diseases may share risk factors with other complex diseases. However, most of the published studies focused on identifying risk exposures for one disease or several diseases and virtually no studies have been conducted to systematically evaluate risk exposures for all common digestive diseases and examine whether these exposures are causally associated with digestive diseases, whether digestive diseases will result in other complex diseases, and whether the relationships between risk exposures and other complex diseases are mediated through digestive diseases. In this project, we aim to comprehensively investigate the causes and consequences of common digestive diseases. To address these issues, we will perform a prospective observational study, a phenome-wide association study, and a mendelian randomization analysis using data from the UK Biobank. We will also develop risk prediction models for digestive diseases and evaluate the potential interactions between environmental and genetic factors on the risk of digestive diseases. We will start analyses as soon as data are available and plan to finish this project and send manuscripts to authors for review within 36 months after we receive the data. We hope that our study will provide a profile of the causes and consequences of digestive diseases and may help identify novel biological pathways and therapeutic targets for improving prevention and treatment of digestive diseases and other complex diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causes-and-consequences-of-fat-distribution-patterns-in-the-liver-pancreas-skeletal-muscle-heart-kidney-and-visceral-bed

Causes and consequences of fat distribution patterns in the liver, pancreas, skeletal muscle, heart, kidney, and visceral bed

Last updated:
ID:
93426
Start date:
22 February 2023
Project status:
Current
Principal investigator:
Dr Hajime Yamazaki
Lead institution:
Kyoto University, Japan

Obesity is defined as the accumulation of excessive fat. Fat can accumulate in various organs in the human body, including the liver, pancreas, muscle, kidneys, heart, and visceral bed. There are substantial individual differences in fat accumulation patterns. Based on the fat content in the liver, pancreas, muscle, and visceral bed, we recently identified four specific patterns of fat distribution, each associated with a different diabetes risk: liver fat pattern (mainly liver fat and visceral fat), pancreatic fat pattern (mainly pancreatic fat, visceral fat, and muscle fat), muscle fat pattern (only muscle fat), and low fat pattern (low fat in organs). However, we have not obtained sufficient data on whether these fat distribution patterns can be extrapolated to large populations, what potential health-related events (e.g., diabetes, liver diseases, and pancreas diseases) these fat distribution patterns lead to, why individual differences in fat distribution occur, and how adding fat content to other organs, such as the heart and kidneys, changes the fat distribution patterns . Fat in the organs can be an indicator of organ quality, while organ volume reflects organ quantity; therefore, we would evaluate the effects of both fat and organ volume in our project. We will address the above questions using UK Biobank data, including imaging and genetic data, physical measures, laboratory data, questionnaires, and health-related event data. During our project, with an expected duration of 36 months, we believe that we could answer the above questions. The generated data and evidence from our project would substantially contribute to personalized recommendations using imaging modalities for the prevention of lifestyle-related diseases, such as diabetes.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causes-and-consequences-of-human-trait-variation

Causes and consequences of human trait variation

Last updated:
ID:
116122
Start date:
12 January 2024
Project status:
Current
Principal investigator:
Professor Peter Visscher
Lead institution:
University of Oxford, Great Britain

Humans vary tremendously across a wide range of traits, for example in colour, height, weight, blood pressure, and behaviour. Some of that variation is due to genetic effects or captured by known environmental factors. However, we still know very little about how genetic variation between people combined with their environmental exposures lead to differences in outcomes in life, including risk of common diseases such as cardiovascular disease, cancer and disorders of the brain, psychological and economic well-being and longevity. We aim to use genomic, trait and disease data in the UK Biobank to develop and apply statistical approaches to address questions about how genomic variation causes individual differences. The rationale for this approach is what we know that the DNA is essentially unchanged during a person’s life and comes before a person is exposed to the environment.

We will use associations between DNA variants and knowledge about the genome to identify variants and genes that cause individual differences; develop and apply statistical approaches to use all genomic and trait data simultaneously to quantify the relationships between traits and test the limits of predicting an individual’s risk of disease in the future; develop new phenotypes from existing genomic and trait data, quantify their genetic basis and how they are correlated with disease and other traits. These aims will take at least 3 and probably 6 years to address.

Understanding the genetic basis of relationships between molecular, physiological and behavioural phenotypes and life outcomes is of considerable research and public health interest. It will lead to a better understanding of the genetic factors and biochemical pathways underlying individual differences, including those for common diseases. The impact of this research will be new knowledge about factors that causes disease and adverse effects in life. Such knowledge may lead to new prevention or treatment strategies. Another impact will be the distribution of computer programs that other researchers can use in different datasets.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causes-and-consequences-of-valvular-heart-disease

Causes and consequences of valvular heart disease

Last updated:
ID:
22207
Start date:
24 November 2016
Project status:
Current
Principal investigator:
Professor Kazem Rahimi
Lead institution:
University of Oxford, Great Britain

Valvular heart disease refers to a group of conditions that denote damaged or defective heart valves. Mitral and aortic valve disease tend to be more commonly diagnosed and have more serious health consequences. In an aging population, the burden of valvular heart disease is increasing. However, a large proportion of valvular heart disease is still considered to be ‘degenerative’ with no clear understanding of its causes and no established preventative strategies. In this application, we propose to make use of the multi-modal data from the UK Biobank to investigate the causes and consequences of valvular heart disease. Better understanding of underlying causes is the first critical step in developing interventions for prevention of valvular heart disease and its progression. – We first report the incidence and prevalence of valvular heart disease in this contemporary population. The cases will be identified as reported in electronic health records (EHR) and further complemented by cardiac MRI and echocardiography studies that are ongoing.
– We then predict the risk of developing individual types of valvular disease. Modelling will take account of potential interactions by other vascular diseases, in particular when EHR data are used.
– We will complement the analysis by conducting a series of Mendelian randomisation studies to identify potential causal pathways for disease development and progression.
The full cohort, plus imaging sub-study cohort, and access to derived genetic markers.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causes-correlates-and-consequences-of-reduced-renal-function

Causes, Correlates and Consequences of Reduced Renal Function

Last updated:
ID:
31852
Start date:
2 March 2015
Project status:
Closed
Principal investigator:
Professor Adam Butterworth
Lead institution:
University of Cambridge, Great Britain

We intend to investigate the potential causes, correlates and consequences of reduced renal function and chronic kidney disease (CKD). This project aims:

(1) To understand the characteristics of self-reported CKD patients.

(2) To assess the cross-sectional associations of renal biomarkers with biological, lifestyle and other characteristics.

(3) To examine the within-person variability of renal biomarkers.

(4) To identify genetic determinants of CKD and renal biomarkers.

(5) To characterise the associations of CKD and impaired renal function with the risk of several diseases. CKD is a major global health problem affecting about 15% of the adult population worldwide. This research will provide a comprehensive assessment of the causes, correlates and consequences of impaired renal function and CKD, and will give insight into possible therapeutic areas, which could be investigated further in the view of developing future medication. In the first phase, we will perform analyses investigating the prevalence of self-reported CKD and its associations with other prevalent diseases as well as with socio-demographic, lifestyle, environment, early life, psychosocial and physical measures. When available in UK Biobank, we will analyse the genetic and biochemical data to assess the genetic determinants, cross-sectional correlates and within person-variability of markers of renal function. Once suitable numbers of disease outcomes have accrued we will examine the relationships of impaired renal function with future health outcomes. The full cohort will be required.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causes-of-biliary-tract-cancers-evidence-from-prospective-observational-studies-genome-wide-association-analysis-and-mendelian-randomization

Causes of biliary tract cancers: evidence from prospective observational studies, genome-wide association analysis and mendelian randomization

Last updated:
ID:
45902
Start date:
26 November 2018
Project status:
Current
Principal investigator:
Professor Ben Zhang
Lead institution:
Sichuan University, China

Biliary tract cancers are a clinically heterogeneous group of uncommon cancers including gallbladder cancer, cholangiocarcinoma, and ampulla of Vater cancer. The pathogenesis of these highly lethal cancers is poorly understood. To date, only a few environmental and genetic factors have been identified to be associated with biliary tract cancers. Furthermore, environmental factors such as gallstones are strongly associated with biliary tract cancers, particularly one of the specific subtypes, but whether these associations are causal remains unclear. This question has clinical significance since prevention or early treatment of gallstones may ultimately reduce the incidence of biliary tract cancers. In this project, we aim to systematically investigate environmental and genetic factors of biliary tract cancers, and examine whether risk factors identified in prospective observational studies such as gallstones, cholecystitis and obesity are causally associated with risk of biliary tract cancers. To address these issues, we will conduct a meta-analysis of prospective observational studies, a genome-wide association study and a mendelian randomization analysis using data from UK Biobank and other sources. We will start analyses as soon as data are available and plan to finish this project and send manuscripts to authors for review within 18 months after we receive the data. We hope that this study will provide summary data for the relationships between environmental and genetic factors and risk of biliary tract cancers, and advance our understanding of the etiology of these uncommon but highly lethal cancers. Our study is consistent with the goal of UK Biobank that dedicates to improve the prevention, diagnosis and treatment of a wide range of serious and life-threatening illnesses like biliary tract cancers.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causes-of-individual-differences-in-cognitive-and-mental-health

Causes of individual differences in cognitive and mental health

Last updated:
ID:
16406
Start date:
15 February 2016
Project status:
Current
Principal investigator:
Professor Danielle Posthuma
Lead institution:
VU University Amsterdam, Netherlands

The main goal of our study is to quantify and understand the role of genetic variants, the environment (including lifestyle), and their interaction on outcomes related to cognitive health. In doing so we will combine expertise of statistical genetics, medical genetics, bioinformatics and functional genomics. We are specifically interested in the following health-relevant outcomes from the U.K. Biobank data: cognitive function (incl. normal function and dementia), mental health (incl. depression, neuroticism, personality, smoking, and alcohol drinking), and brain MRI. Our research will contribute to quantifying and understanding how several risk factors (e.g. lifestyle, environment, genes), both separately and in combination, influence cognitive health as well as the comorbidities between different cognitive health outcomes. Our study will consist of a combination of methods, including:
– Genome-wide association studies (GWAS) that aim to identify individual genetic variants associated with a particular outcome.
– Comorbidity analyses, using e.g. meta-analytic techniques, LD score regression or BOLD-GREML methods to quantify the extent of genetic overlap between particular outcomes
– Gene-set analyses (e.g. using MAGMA and INRICH tools) and bioinformatic secondary analyses to understand genetic findings in terms of their biological function
– Heterogeneity analyses to determine genetic subgroups of individuals
– Annotation of genetic findings using external information from e.g. expression or quantitative proteomics data
– Gene-by-environment correlation and interaction analyses to quantify the relevance of the interplay between genes and environment (including lifestyle) on outcomes related to cognitive health We aim to use all available observations in the UKB that are currently released and will be released in the future, and that have been successfully genotyped and have measures of relevant outcomes. ?


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/causes-risk-prediction-and-outcomes-of-gastrointestinal-diseases

Causes, risk prediction and outcomes of gastrointestinal diseases

Last updated:
ID:
551355
Start date:
30 April 2025
Project status:
Current
Principal investigator:
Dr Shaohua Xie
Lead institution:
Karolinska Institutet, Sweden

Gastrointestinal diseases, including the deadly gastrointestinal cancers and related benign conditions, are huge disease burden globally. The causes of theses diseases are not completely understood, and the current prevention and treatment strategies remain to be improved. With the use unique comprehensive data from a large number of participants in UK Biobank, we aim to explore the genetic and non-genetic causes of gastrointestinal diseases, and also examine gene-environment interaction in the development of these diseases. We will develop and validation risk prediction models for these diseases which may be helpful for tailored prevention, early detection and treatment. We will also investigate the factors influencing the outcomes of these diseases. The research methodologies involve observational studies, Mendelian randomization analysis, as well as phenome- and genome-wide association analyses. The research findings will provide valuable scientific evidence supporting improved prevention and treatment of gastrointestinal diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/ccl3l1-and-ccr5-delta-32-cnv-study-in-extremes-of-the-lung-function-distribution-in-smokers

CCL3L1 and CCR5 delta 32 CNV study in extremes of the lung function distribution in smokers

Last updated:
ID:
7140
Start date:
1 September 2014
Project status:
Closed
Principal investigator:
Professor Edward Hollox
Lead institution:
University of Leicester, Great Britain

COPD is a long-term progressive condition affecting ~900,000 people in the UK accounting for ~30,000 deaths and over £500m of NHS costs annually. Copy number variation (CNV) could account for some of the variability in COPD risk and lung function. We aim to study CNV of two genes in relation to COPD and lung function. CCL3L1 encodes a pro-inflammatory cytokine. There is a relationship between CCL3L1 copy number (CN) and protein expression levels, and some evidence that CCL3L1 CN is associated with lung function. CCR5 encodes a receptor for CCL3L1 and contains a polymorphic 32bp deletion. Genetic approaches to understanding the mechanisms underlying COPD aim to discover targets for drug development and unravel disease heterogeneity facilitating stratified approaches to treatment. CCL3L1 and CCR5d32 CN measurements for all UK Biobank particpants included in this project will be returned to UK Biobank to be made available to other researchers. CN at some regions can be inferred from SNP data but complex CN regions (where CN varies from 0 to 6 or more) require custom-designed assays. CCL3L1 CN varies from 1 to 6 and the ?gold standard? approach for measurement is the paralogue ratio test (PRT). CCL3L1 is a ligand for CCR5, a receptor that contains a polymorphic deletion associated with other diseases, including asthma. We will genotype CCL3L1 CN and CCR5d32 in smokers with good or bad lung function and test for association with lung function to clarify the therapeutic opportunities of the CCL3L1-CCR5 axis in COPD. We will genotype 2500 heavy smokers with good lung function and 2500 heavy smokers with poor lung function. These will be selected from the 50,000 individuals for whom DNA has been extracted for the UK BiLEVE consortium.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/central-and-peripheral-sympathetic-nervous-system-activity-in-older-adults-feasibility-study-on-dpuk-cohorts-to-obtain-preliminary-data-pilot-project-funded-by-early-career-researcher-dpuk-grant

Central and peripheral sympathetic nervous system activity in older adults: feasibility study on DPUK cohorts to obtain preliminary data (Pilot project funded by Early Career Researcher DPUK grant)

Last updated:
ID:
54410
Start date:
6 March 2020
Project status:
Current
Principal investigator:
Dr Grazia Daniela Femminella
Lead institution:
Imperial College London, Great Britain

Alterations of the so called “autonomic nervous system” are common in patients with dementia. In particular, a region of the brain called locus coeruleus (LC) is the major centre in regulating autonomic function, as well as several cognitive responses. Because of its small size, it has been difficult to evaluate the LC structure and function. Nevertheless, recent advances in magnetic resonance imaging (MRI) techniques, have made it possible to visualize this structure. However, few studies have so far specifically looked at LC changes in patients with Alzheimer’s disease. The availability of large imaging UK cohorts, such as UK Biobank, make it possible to test whether structural and functional MRI data can be obtained on currently available images.
The aim of this proposal is to evaluate whether structural and functional MRI data for the LC can be obtained, using available protocols, and if those are related to heart rate variability, a measure of autonomic nervous system function, which can be obtained from ECG data in the same cohort. Establishing whether structural and functional LC measures can be obtained on existing data would be extremely important to test hypothesis on LC involvement in aging and in AD pathology.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/central-nervous-system-adaptation-in-ankle-instability

Central nervous system adaptation in ankle instability.

Last updated:
ID:
62721
Start date:
25 August 2020
Project status:
Current
Principal investigator:
Dr Xiao'ao Xue
Lead institution:
Fudan University, China

Lateral ankle sprain is one of the most common injury in sport, and 40% of patients would have persistent symptoms and develop into chronic ankle instability (CAI). Sensorimotor deficit was thought to be a reason for the symptoms of joint instability, and functional changes of central nervous system had been found in CAI patients according to previous electroencephalogram studies.

In this study, we aimed to use the Brian MRI data from UK biobank to investigate the potential structural and functional changes of central nervous system caused by ankle joint instability. T1 structural, diffusion and resting-state functional MRI would be evaluated, and the comparison would be performed between healthy controls, patients with ankle instability alone and patients with both ankle instability and recurrent sprains.

This research would explore the mechanism of central nervous system adaptation in ankle instability and promote the development of more targeted balance rehabilitation projects for CAI patients.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cerebral-correlates-of-the-apoe-and-other-genetic-risk-factors-for-alzheimers-disease-in-healthy-middle-aged-individuals

Cerebral correlates of the APOE and other genetic risk factors for Alzheimer’s disease in healthy middle-aged individuals

Last updated:
ID:
31085
Start date:
3 July 2019
Project status:
Closed
Principal investigator:
Dr Natalia Vilor-Tejedor
Lead institution:
Barcelonabeta Brain Research Center, Spain

The e4 allele of the APOE gene is associated to a higher risk of Alzheimer?s disease (AD). Cognitively healthy APOE-e4 carriers have been reported to display lower gray matter volume in brain regions known to be affected by AD such as the hippocampus. Yet, numerous studies have not been able to detect any significant differences or even found areas of greater grey matter volume in e4 carriers as compared to non-carriers. We aim at investigating the cerebral neuroimaging patterns associated to the APOE genotype in healthy middle-aged subjects. Converging evidence supports that Alzheimer?s disease has a long and protracted preclinical stage. The earliest cerebral alterations are thought to happen even 20 years before the onset of the symptoms. We aim at studying cerebral patterns of individuals with an increased genetic risk of developing AD in order to better characterize this preclinical stage and to reveal any potential interactions with other modifiable risk factors (hypercholesterolemia, hypertension, elevated body-mass-index, etc?) which have also been linked to the APOE-e4 genotype. We could therefore provide with novel evidence supporting primary prevention of AD. In healthy individuals, we will analyze brain regions known to be affected in AD and will seek for the cerebral fingerprint of the APOE genotype. We will also seek for associations between these brain regions and other factors also known to be affected by the APOE genotype such as high blood cholesterol among other factors known to increase the risk of both AD and cardiovascular disease. This investigation might help us understanding the underlying biological factors involved in APOE increasing the risk of these conditions. We would like to include all available individuals with a minimum of a T1-weighted magnetic resonance images and a genome-wide analysis (to obtain the APOE genotype). Additional analyses would also require other magnetic resonance image modalities and variables such as hypertension, hypercholesterolemia, body-mass-index, dietary and lifestyle questionnaires, as well as psychometric scales.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cerebral-small-vessel-disease-and-alzheimers-disease-susceptibility-a-genome-wide-interaction-study

Cerebral small vessel disease and Alzheimer’s disease susceptibility: a genome-wide interaction study

Last updated:
ID:
56197
Start date:
1 September 2020
Project status:
Current
Principal investigator:
Dr Walter Swardfager
Lead institution:
Sunnybrook Research Institute, Canada

Alzheimer’s disease (AD) is the commonest cause of dementia, in which patients will experience a progressive decline in cognition, language, memory, and physical ability. People with dementia due to AD in life are typically found to have multiple additional pathologies at autopsy. In particular, cerebral small vessel disease (SVD), is one of the commonest in the majority late-onset cases of AD (the more common form of AD and typically begins in patients age 65 and older). Cerebral SVD is a type of vascular lesion in the brain, with certain characteristic impairments in brain structures that can be assessed via magnetic resonance imaging (MRI). On the other hand, conventional genome-wide association studies (GWAS), a type of genetics analysis that surveys the whole genome for genetic variants associated with certain disease or physical traits, revealed ~21 genetic variants associated with AD. However, mechanisms underlying AD onset and progression remain unresolved. Conventional GWAS also focus on genetic variants with main effects and often eliminate variants that are likely to show conditional effects. A novel approach is to conduct a genome-wide interaction study (GWIS) which focuses on important interaction effects. Here, I will survey the human genome for interactions between SVD brain imaging and genetic characteristics, which contribute together to increase the risk of AD diagnosis, and to AD-associated brain imaging and cognitive disease characteristics. We aim to conduct this GWIS over the course of 3 years, in which we will conduct quality control and sophisticated statistical analyses for the anticipated massive datasets of brain imaging, cognitive, and genetic data. The pathological heterogeneity in AD has posed a tremendous challenge in developing treatments for the disease. Via analyzing the above mentioned data variables in individuals with normal cognitive status and demented populations in the UK Biobank, our goal is to disentangle the roles of heterogeneous pathological factors underlying the clinical symptoms in order to overcome the tremendous challenge this poses in the quest for effective treatments.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cerebrovascular-mapping-using-human-resting-state-functional-mri-data

Cerebrovascular mapping using human resting-state functional MRI data

Last updated:
ID:
151378
Start date:
17 April 2024
Project status:
Closed
Principal investigator:
Dr Christopher Murray
Lead institution:
Voxel AI Inc., Canada

Cerebrovascular reactivity (CVR), an index of the brain’s vascular response, has been shown to provide useful information in diagnosing and treating patients with different brain pathologies, as well as mapping the neurovascular effects associated with normal aging. Commonly, CVR mapping is performed using hypercapnic (HC) gas inhalation during functional MRI (termed HC-CVR), but the cost and complexity associated with using the inhalation equipment, as well as the inability of some patients to tolerate the protocol, has presented a major hindrance to the wide-scale application of CVR across MRI centres and hospitals. To circumvent these practical challenges, recent studies have used functional MRI data easily collected during natural breathing to map CVR and have shown that many aspects of CVR can be reliably estimated when the subject is simply laying at rest (called resting-state CVR or RS-CVR). The overarching goal of the current proposal is to compare different established RS-CVR metrics, as well as explore new RS-CVR metrics that can also be used to assay HC-CVR. To provide a practical test of these different metrics, we assess their ability, via a machine learning model, to predict subject brain age using publicly available data sets. Together, this approach promises to provide new data and knowledge that will have practical benefit for both the academic community and health care industry. Indeed, due to the simplicity of this approach and its inexpensive implementation, RS-CVR has the potential to serve as a much more wide-spread clinical and research tool. This project will occur over an approximately 3-year time period.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/change-in-body-composition-gut-microbiota-and-modifiable-risk-factors-in-healthy-aging

Change in body composition, gut microbiota, and modifiable risk factors in healthy aging

Last updated:
ID:
1042056
Start date:
11 October 2025
Project status:
Current
Principal investigator:
Dr Yuanyue Zhu
Lead institution:
Ruijin Hospital, China

Age-related changes in body composition are fundamental determinants of healthy aging trajectories. Concurrently, aging processes and adipose redistribution can both contribute to gut dysbiosis. While these biological changes are inevitable, modifiable factors including lifestyle behaviors and socioeconomic determinants may significantly influence these trajectories and their health consequences. Understanding how modifiable factors interact with body composition changes and gut microbiota is crucial for developing targeted interventions that promote successful aging.
Therefore, this investigation aims to examine the complex interplay between longitudinal body composition dynamics (fat mass, lean mass and mineral density), gut microbiota profiles, and health outcomes including cancer, cardiovascular disease, and mortality within the UK Biobank cohort.
Research Objectives
(1) characterize age-related trajectories of fat mass, lean mass, and bone mineral density across different demographic groups
(2) identify modifiable factors (including lifestyle behaviors and socioeconomic status) and gut microbiota signatures associated with favorable compositional maintenance
(3) evaluate the combined effects of modifiable factors, body compositional changes, and microbial diversity on healthy aging
Results will inform evidence-based intervention strategies targeting body composition optimization and microbiome modulation for healthy aging, with implications for precision medicine approaches for aging populations.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/changes-in-resting-state-fmri-and-cognitive-scores-associated-with-sars-cov-2-in-the-uk-biobank

Changes in resting state fMRI and cognitive scores associated with SARS-CoV-2 in the UK Biobank

Last updated:
ID:
98045
Start date:
26 May 2023
Project status:
Current
Principal investigator:
Ms Josefina Weinerova
Lead institution:
University of Nottingham, Great Britain

We aim to address the important question of what are the changes in brain function following Covid-19 infection (also referred to as the long-covid) and how they are associated with cognitive functions such as memory, reasoning or problem-solving.
Previous studies have found that there are differences in brain structure and performance on tasks measuring cognitive function between people who had and had not been infected with Covid-19, and within the same people before and after infection. However, it is yet unclear whether Covid-19 infection further affects the interactions between different brain areas, and how these might correspond to changes in cognitive function. Another unresolved question is whether vaccination prior to infection alleviate the brain changes and cognitive symptoms associated with of long-covid.
We aim to answer these questions in our three-year-long research project. The results of our study could inform the debate concerning the long-lasting effects of Covid-19. More generally, the research into long-lasting effects of infectious diseases can provide necessary data for public health strategies. Additionally, the potential effect of vaccinations on severity of long-covid symptoms could likewise inform policy making regarding access to and promotion for Covid-19 vaccines in the future.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/changing-the-sodium-homeostasis-paradigm-the-role-of-macrophages-in-sodium-sensitivity-a-candidate-gene-study-using-the-uk-biobank-cohort

Changing the sodium homeostasis paradigm: the role of macrophages in sodium sensitivity, a candidate gene study using the UK biobank cohort.

Last updated:
ID:
263250
Start date:
17 December 2024
Project status:
Current
Principal investigator:
Dr Jacob Murray
Lead institution:
Amsterdam UMC Research BV, Netherlands

We know that salt intake is important with regards to health. Specifically related to issues such as high blood pressure, heart disease and kidney disease. We are interested in figuring out how and why some people have higher blood pressure when they eat a high salt diet and why others do not. Observations on patients with diseases related to the immune system or treated with immune suppressing drugs have suggested that the immune system might be involved in the causes of high blood pressure. Scientists have developed mice which have high blood pressure when they are fed too much salt. Research on such mice has found that if we interfere with parts of the immune system we can influence the effect salt has on their blood pressure.

We hope to identify genetic variation related to the activity of the immune system in humans to see if the same parts of the immune system also affect how a patient’s blood pressure responds to salt intake. We hope to identify this genetic variation in the UK biobank data, before seeing if we can find the same variation in other datasets to attempt to confirm the validity of our initial findings. If we are confident we have identified such genetic variants, it will help us carry out other experiments to better confirm and describe how salt affects the immune system and how these effects influence high blood pressure and disease.

Our expectation is that this process will take approximately 12 months with time spent analysing the data and comparing results with other datasets. We hope that our research, when combined with other research into what causes high blood pressure, targeted treatments can be developed aimed at treating the specific causes for blood pressure in an individual. This kind of personalised medicine is the longer term goal of such research into disease mechanisms.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterisation-of-adme-gene-star-alleles-across-african-ancestry-populations-and-comparison-with-other-global-populations

Characterisation of ADME gene star alleles across African-ancestry populations and comparison with other global populations

Last updated:
ID:
108515
Start date:
14 March 2024
Project status:
Current
Principal investigator:
Dr David Twesigomwe
Lead institution:
University of the Witwatersrand, Johannesburg, South Africa

Background, Rationale, and Aims: Drug response varies across patients-ranging from those who experience treatment failure to those who may suffer adverse drug reactions (ADRs). Genetic variation affecting proteins involved in the absorption, distribution, metabolism, and excretion (ADME) of drugs can considerably alter drug efficacy and contribute to ADRs. This ADME genetic variation is usually represented in the form of star alleles (shorthand for complex variant combinations and structural variants in DNA). An example of ADME genes is cytochrome P450 2D6 (CYP2D6). CYP2D6 metabolises about 20% of commonly prescribed drugs, meaning that variations in CYP2D6 could have a significant impact on the treatment of multiple diseases. CYP2D6 has over 170 star alleles according to research curated by the Pharmacogene Variation Consortium (PharmVar). This includes normal function, decreased function, non-functional, increased function, and unknown function star alleles. It is important to characterise the star allele diversity in ADME genes to lay the foundation for pharmacogenetics testing across clinical settings globally. At present the full catalogue of star alleles in major ADME genes such as CYP2D6 is unknown, while the distribution of known star alleles is also unclear across majority of global populations. The availability of full genomes generated by large consortia and biobanks provides the opportunity to address these gaps in knowledge and to catalyse the implementation of individualised medicine. Our research therefore aims to characterise and compare the genetic diversity in major drug metabolism, drug transporter, and modifier genes across diverse populations represented in the Human, Heredity, and Health in Africa (H3Africa) Consortium data catalogue and the UK Biobank. This study will contribute to identifying potentially novel ADME star alleles that may impact drug response. We are aware that some ADME genes such as CYP2D6 are complex to analyse using standard variant calling bioinformatics tools. We therefore plan to use StellarPGx (pharmacogenomics tool developed by our group) which we specifically tailored to characterising structural variants and identifying novel star alleles in these genes.

Project Duration: 3 years

Public Health Impact: Our proposed study will contribute to characterising the landscape of genetic variation that could impact drug response across populations represented in the UK Biobank and other major data catalogues. Understanding the extent of this pharmacogenomic variation across multi-ancestry cohorts will inform the development of suitable pharmacogenetics testing platforms and guide drug dosing algorithms to promote drug efficacy and minimise the risk of adverse drug reactions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterisation-of-risk-factors-and-their-mechanisms-in-the-development-of-both-common-and-relatively-understudied-conditions

Characterisation of risk factors and their mechanisms in the development of both common and relatively understudied conditions

Last updated:
ID:
67506
Start date:
29 June 2021
Project status:
Current
Principal investigator:
Dr Gillian Reeves
Lead institution:
University of Oxford, Great Britain

Cardiovascular disease (CVD), cancer, chronic and infectious respiratory diseases, and neurodegenerative disorders are the leading causes of death globally and in the UK. Although major risk factors have been identified for some of these diseases, there is limited information regarding the role of other potential risk factors, or on the potential mechanisms underlying these associations, particularly for the less common conditions. We aim to establish which factors are causally associated with disease, elucidate the mechanisms underlying such relationships and identify those individuals at greatest risk of specific diseases. This work will complement and extend the work being carried out in other large cohort studies (such as the Million Women Study and EPIC) and collaborations within the Cancer Epidemiology Unit. The findings from this work and from other multiple large studies within the Unit will contribute to a greater understanding of potentially modifiable risk factors for several common and emerging diseases and allow more targeted screening and therapeutic interventions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterising-and-assessing-long-term-outcomes-of-people-with-cancer-using-real-world-data

Characterising and assessing long-term outcomes of people with cancer using real-world data

Last updated:
ID:
105403
Start date:
19 December 2023
Project status:
Current
Principal investigator:
Dr Danielle Newby
Lead institution:
University of Oxford, Great Britain

The aim of this research is to use medical records, information about lifestyle and information from people genes from UKBiobank to understand more about patients with cancer and how this impacts their future health and survival after diagnosis.

The number of people diagnosed with cancer is increasing each year. Cancer is the leading cause of death in the UK and globally. The increasing numbers increase costs and the pressure on healthcare systems. There has been great advancement in treatments for many cancers however clinical trials are costly and take time to complete. We can use data already collected such as data from UKBiobank to provide answers about cancer patients, their treatments and survival.

Using UKBiobank we will determine what diseases, medications and lifestyle factors patients with cancer had before they were diagnosed. This will help identify risk factors that can be used to predict how long cancer patients live after diagnosis. We will use information on cancer treatments to compare which ones have better survival for patients and use information from people genes to identify genes that may have better or worse outcomes for patients.

For this project, we will use a variety of statistical methods and machine-learning approaches. It is important to use different methods to increase confidence in results by giving a complete picture of cancer patients which can lead to better treatments and ultimately benefit patients.

This project is in the public interest as cancer research is crucial to improve the prevention, detection and treatment of different cancers, and ensure that patients live longer and better-quality lives. The project duration plans to take at least 36 months.!


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterising-blood-microbes-in-health-and-disease

Characterising blood microbes in health and disease

Last updated:
ID:
144825
Start date:
5 December 2023
Project status:
Current
Principal investigator:
Dr Robin Mjelle
Lead institution:
Norwegian University of Science and Technology, Norway

Microbes such as bacteria and viruses are important players in disease and health. It is unclear what role blood microbes play and if they can be utilised as biomarkers and reflect underlying disease. In this study we will investigate if blood samples from the UK biobank contain microbes and if the presence of these microbes correlate with infectious diseases and cancer. The study will gain knowledge on the general landscape of microbes in the blood but also disease-specific microbes that can predict future disease such as cancer. The project will last for three years starting in 2024.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterising-imaging-and-clinical-features-of-normal-pressure-hydrocephalus-in-the-uk-biobank

Characterising imaging and clinical features of normal pressure hydrocephalus in the UK Biobank

Last updated:
ID:
654960
Start date:
14 April 2025
Project status:
Current
Principal investigator:
Dr Clara Belessiotis-Richards
Lead institution:
King's College London, Great Britain

Scientific rationale:

Normal pressure hydrocephalus (NPH) is a potentially treatable condition that causes gait disturbance, urinary incontinence, and dementia. Diagnosis of NPH is primarily based on brain imaging and clinical features. Treatment is through insertion of a ‘shunt’, which improves symptoms and prolongs independence. However, NPH may be under-diagnosed, as international community surveys have found higher than expected rates of imaging and clinical features of NPH in the general population. Estimates suggest that 0.5-2% of the general population aged 60 years and above may have NPH. Imaging features of NPH include ventriculomegaly, narrowed callosal angle, and white matter hyperintensities. Automated brain imaging measures might have potential to identify NPH. However, little is known about the prevalence of NPH clinical and imaging features in the UK. With rising dementia rates globally, increasing our understanding of treatable conditions such as NPH is key.

Objectives:

In this study, we seek to characterise the period prevalence of imaging features of NPH in the UK Biobank for the first time, and describe their association with symptoms suggestive of NPH (eg. gait disturbance) and with diagnosis of NPH. We will describe how these imaging features vary according to demographic, clinical, symptoms, and risk factors and whether they are associated with outcomes, such as mortality. We plan a predominantly descriptive study, as NPH is a rare outcome. To do this, we will use pre-existing brain segmentation data and run additional data processing programs on raw brain MRI images in the UK Biobank, and compare these with clinical data.

Research questions:

What is the period prevalence of imaging biomarkers of NPH, as listed above, in people aged 60 years and above in the UK Biobank?
Are these imaging features associated with diagnosis of NPH?
How do these imaging features vary according to factors?


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterising-multimorbidity-and-frailty-associated-with-cardiometabolic-disease

Characterising multimorbidity and frailty associated with cardiometabolic disease

Last updated:
ID:
59585
Start date:
30 March 2020
Project status:
Closed
Principal investigator:
Dr Richard Cubbon
Lead institution:
University of Leeds, Great Britain

Advances in disease prevention and treatment mean that our society is rapidly aging and people are often living with multiple long-term diseases, a phenomenon called multimorbidity. Disease and the normal ageing process can reduce the ability of key body systems to respond to the day to day stresses we encounter; for example people with heart failure and also the very elderly are less able to tolerate strenuous exercise or an infection. This reduced tolerance of stress is sometimes referred to as frailty, and is associated with poorer quality of life, increased healthcare utilisation and reduced survival rates. Multimorbidity does not develop at random, but instead is more likely to come in clusters, such as people with heart failure also having diabetes. This accumulation of diseases is linked to greater change of frailty, but the relationship between common disease clusters and frailty is not well explored. The UK Biobank dataset allows an opportunity to address these knowledge gaps, with potential implications for understanding of disease clustering and its impact on people and society.

Due to our expertise in the field, we aim to focus on common cluters of cardiovascular (like heart failure and high blood pressure) and metabolic diseases (like diabetes or obesity) in a project that is expected to last 3 or more years. We intend to identify important clusters of these diseases and also explore how they link with frailty. We hope to identify risk factors for developing these clusters, understand better what defines and differentiates the clusters, and observe how these groups of people progress in future in terms of health care use and long-term survival. It is hoped that these data may help to guide preventative and therapeutic approaches for individuals and inform public health and social care policy.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterising-neural-network-markers-of-occupational-wellbeing

Characterising neural network markers of occupational wellbeing

Last updated:
ID:
62188
Start date:
29 September 2020
Project status:
Closed
Principal investigator:
Dr Charlotte Rae
Lead institution:
University of Sussex, Great Britain

Our employment status and what we do for a living can have numerous impacts on our wellbeing and mental health, from under- or over-employment, to shift work, and time spent commuting instead of with our families and relaxing. However, the brain mechanisms by which such occupational factors influence our wellbeing, and make us vulnerable to experiencing poor mental health, are less well understood. This means that we do not fully understand why some people become stressed or unwell as a result of occupational issues, while others remain unaffected.

This project aims to find out what aspects of brain function are associated with various occupational measures, such as employment status, hours worked, shift work, and commuting. The UK Biobank database contains information on these occupational measures, as well as participants’ wellbeing and mental health, and MRI brain scans. This project will use functional magnetic resonance imaging (fMRI) measures of brain activity, recorded while participants undertook an emotional face perception task, and also while they rested quietly during brain scanning. The research team will analyse the data to look for patterns of brain activity that are more common in people with certain types of employment status, working long hours, or doing shift work. This will tell us whether various patterns of working tend to be associated with certain patterns of brain activity.

Then, statistical tests will be conducted to see if these patterns of brain activity are a likely causal factor in the impact that people’s employment has on their wellbeing and mental health. This will help us identify which sub-groups of people are likely to be neurologically vulnerable to work-related wellbeing and mental health problems.

These results will be of interest to employers who want to keep their workforce healthy and productive; to employees who want to consider making requests to their employer for better wellbeing (such as reduced hours); and to policymakers who want to support optimum economic and public health.

The project is being conducted by a team of researchers at the University of Sussex, including Principal Investigator Dr Charlotte Rae and PhD student Raul Ungureanu. The project will form part of Raul’s PhD thesis on the neural mechanisms of wellbeing in the workplace. As a result, the project duration (36 months) matches the duration of Raul’s PhD at Sussex, so we expect to be able to share the project’s results in approximately 2023.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterising-plasma-proteomic-profiles-in-over-50000-uk-biobank-participants-for-better-understanding-of-protein-variability-and-associations-in-health-and-disease

Characterising plasma proteomic profiles in over 50,000 UK Biobank participants for better understanding of protein variability and associations in health and disease

Last updated:
ID:
94598
Start date:
14 December 2022
Project status:
Current
Principal investigator:
Dr Ida Grundberg
Lead institution:
Olink Proteomics AB, Sweden

It is well known that many of the treatments and procedures available for patients in medicine today are effective only for a subset of the patients that are targeted. Precision medicine is an emerging concept aiming to more precisely identify whether a patient is suitable for receiving a given treatment, which would benefit the patient by ensuring they get the right treatment and reducing the risk of unnecessary side effects, in addition to lower the cost of public health care. To achieve this, new and improved diagnostic tools are needed to identify and classify disease early and predict whether a treatment is suitable for an individual patient. Large studies of protein measurements in the population are an essential step towards achieving this goal and is done in the field of proteomics. Such studies can identify proteins that change with disease and reflect biology in real time.

Recent technological advancements have enabled the measurement of around 1,500 proteins in the blood of more than 50,000 UK Biobank participants through the UK Biobank Pharma Proteomics Project (UKB-PPP) initiative. This is the first opportunity for large scale proteomics research and makes UK Biobank an essential reference resource. In this project, which is expected to be completed within three years, we aim to accelerate the progress towards precision medicine by creating an atlas for protein levels in common and important diseases as well as in health. These results will be available in an open digital platform for the research community to use freely in their research. This initiative will promote scientific progress by allowing researchers over the world to quickly explore and compare protein profiles in health and disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterising-resilience-and-discordance-to-polygenic-risk-across-common-chronic-diseases

Characterising Resilience and Discordance to Polygenic Risk across Common Chronic Diseases.

Last updated:
ID:
1029245
Start date:
28 September 2025
Project status:
Current
Principal investigator:
Dr Xiaoming Xing
Lead institution:
Beijing Hospital, China

Polygenic risk scores (PRS) are valuable for stratifying genetic susceptibility, yet there is often a frequent mismatch between inherited predisposition and observed clinical outcomes. This project will investigate the phenomena of “resilience” (high-PRS individuals remaining disease-free) and “discordance” (low-PRS individuals developing disease). Our objective is to identify protective and risk-enhancing factors by applying a harmonised analytical framework across a wide spectrum of chronic diseases. We will begin by targeting key domains including, but not limited to, cardiometabolic conditions (e.g., type 2 diabetes, cardiovascular disease), respiratory disease (e.g., asthma), and selected cancers. We hypothesise that modifiable factors-including lifestyle behaviours, clinical markers, and molecular profiles-can buffer genetic risk on an additive scale, thereby informing prevention strategies.
For progressive conditions such as diabetic complications or advancing chronic kidney disease, landmark analyses will be applied to assess how exposures measured prior to defined time points shape short-term progression risk. This secondary analysis of UK Biobank will integrate PRS with deep phenotyping from accelerometry, primary care records, NMR metabolomics, and Olink proteomics, linked to outcomes from algorithmically defined fields and national registries.
Analyses will adopt prospective modelling with Cox and competing-risks regression, quantify additive interactions (e.g., RERI, AP), and apply false discovery rate control for multiple testing. To complement these conventional approaches, we will also explore machine learning methods to assist in identifying key multidimensional features.
By systematically characterising resilience and discordance within each disease, this research will advance understanding of the gene-environment interplay, refine communication of genetic risk, and provide evidence to guide targeted prevention and early intervention.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterising-the-causes-and-impacts-of-rare-and-structural-variation-on-human-traits

Characterising the causes and impacts of rare and structural variation on human traits

Last updated:
ID:
100469
Start date:
30 March 2023
Project status:
Current
Principal investigator:
Dr Anjali Hinch
Lead institution:
University of Oxford, Great Britain

The chromosomes we inherit from our parents are not exact copies but mosaics of their chromosomes. These mosaics are created during formation of eggs and sperm when cells cut chromosomes up and re-attach them, sometimes in new combinations, in a process known as recombination. In addition, we carry of the order of ~70 new DNA mutations – changes to our genetic code compared to the DNA inherited from our parents. These mutations can be simple single-base-pair changes or large structural rearrangements of DNA. Together with recombination, mutations ultimately create the genetic diversity we observe in populations world-wide.

This genetic diversity is needed for populations to withstand pathogens and other diseases. However, mutations can themselves be causes of disease. We plan to use the wealth of data generated by the UK Biobank to understand the processes of mutagenesis and their consequences for human phenotypes. We are particularly interested in mutations that occur near DNA breaks, which are an essential part of the process of recombination but can also occur by accident leading to repair-related mutations. A major goal of our work is to understand the mechanisms that generate large structural changes in the genome, many of which are predicted to cause human disease through changes in or loss of gene function. We will link this genetic variation to phenotypes that show significant variation in human populations and to infection outcomes, and we will generate new analysis methods to support this. Many of the biological processes that underpin mutagenesis in the germline also operate during normal cell division within the life of an individual, and so have implications for genetic diseases such as cancers. We will extend our findings to cancer-related phenotypes, such as type of cancer and age at diagnosis. Ultimately, we hope this work will generate new insight into the biological processes underpinning infection and DNA repair-related diseases, with the potential to improve interventions in future.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterising-the-relationship-between-behavioral-and-mental-health-characteristics-and-the-modular-organization-of-the-brain-network

Characterising the relationship between behavioral and mental health characteristics and the modular organization of the brain network

Last updated:
ID:
42826
Start date:
8 November 2018
Project status:
Current
Principal investigator:
Dr Chadi Abdallah
Lead institution:
Baylor College of Medicine, United States of America

We will investigate how the organization of the brain connectome (i.e., network of neural connections) is linked to the behavior. More specifically, we will use concurrent neuroimaging and behavioral phenotype data to identify meaningful “clusters” that can be identified across these two sets. By applying these analyses to the diverse population of the UK Biobank, we will be able to identify clusters of linked brain-behavior profiles (“biotypes”), including some that may represent states of psychopathology. These biotypes may be more representative of the underlying neurophysiology compared to traditional constructs based that are behavior alone (e.g., Diagnostic and Statistical Manual of Mental Disorders [DSM]). Additionally, this process will be applied recursively and will allow the identification of “sub-biotypes” (e.g., subtypes of depression). By characterizing these biotypes, clinically-relevant findings may find potential use in the clinic as biomarkers of psychopathology. Longitudinal data from the UK Biobank will allow us to tease out whether biotypes have differential temporal trajectories (e.g., which biotypes are at risk of significant neuropsychiatric morbidity), and whether they can be used pragmatically to predict such outcomes.

The analyses will be conducted on all individuals who have available neuroimaging data, and we estimate that the project will be complete by 24 months from the start date.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterising-the-role-of-lhx9-in-infertility-and-disorders-of-sex-development

Characterising the role of LHX9 in infertility and disorders of sex development.

Last updated:
ID:
110462
Start date:
13 October 2023
Project status:
Current
Principal investigator:
Professor Megan Wilson
Lead institution:
University of Otago, New Zealand

Infertility, which is defined as the inability to fall pregnant after 12 months of regular unprotected intercourse, affects 1 in 6 individuals over the course of their lifetime. The inability to fall pregnant can be distressing for couples and cause them to face stigmatisation in the wider community. While assisted reproductive technologies, such as in vitro fertilisation (IVF), are helping infertile couples to conceive, there is still a lack of understanding of the underlying biological cause of infertility in many individuals. An improved understanding of the genetic causes of infertility could improve someone’s chances of naturally conceiving a child or aid in developing drugs or medical techniques to improve their fertility.
LHX9 is a gene known to be involved in the formation and function of the ovaries and testes and when this gene is absent in mice, they do not develop ovaries or testes. There is some evidence that mutations in this gene could be causing issues with the development and function of the reproductive organs in humans, including their fertility. However, there are not enough examples to confirm this theory. This project will use the genetic data from the UK Biobank and health information about people’s fertility history to identify other examples of genetic mutations in LHX9 to show its role in human infertility. This research is part of a Ph.D. project, so it is expected to take 3 years. It hopes to provide


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterization-of-actionable-pharmacogenetic-pgx-variation-across-population-based-cohorts

Characterization of actionable pharmacogenetic (PGx) variation across population-based cohorts

Last updated:
ID:
69407
Start date:
1 February 2021
Project status:
Current
Principal investigator:
Dr Daniel Rotroff
Lead institution:
Cleveland Clinic Foundation, United States of America

The field of pharmacogenetics (PGx) focuses on understanding the role that genetics plays related to individual responses to medications. The identification and characterization of clinically relevant genetic variants has great potential to enhance clinical benefit, decrease adverse drug reactions, and lower the cost of treatment by optimizing drug selection and dosing for an individual. However, incorporating genetic information into regular clinical practice has seen limited uptake. Furthermore, existing research efforts are often challenged by small sample sizes or they lack of rich clinical data on disease and medications. Here, our motivation is to characterize the occurrence of key genetic variants and determine whether they are associated with increased medication or dosing changes in electronic medical records systems. Ultimately, this research will improve the understanding of the how PGx testing may influence clinical decision making in a real-world setting and may lead to personalized prescriptions with improved medication response rates and fewer the adverse drug effects.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterization-of-functional-and-morphological-mri-features-in-subjects-with-bdnf-val66met-polymorphism

Characterization of functional and morphological MRI features in subjects with BDNF Val66Met polymorphism .

Last updated:
ID:
40157
Start date:
22 February 2019
Project status:
Closed
Principal investigator:
Mr Julio Plata-Bello
Lead institution:
Hospital Universitario de Canarias, Spain

Brain Derived Neurotrophic Factor (BDNF) is a protein that plays an essential role in regulating brain functioning. A polymorphism of the BDNF gene (Val66Met) has been reported as affecting the functioning of this protein. Bearing this mind, this polymorphism has been associated to the development of neuropsychiatric disorders, schizophrenia or Alzheimer disease. Furthermore, this polymorphism has also been reported to have an association with the response of antidepressant treatments and with the evolution of neurodegenerative diseases.
Some studies have reported differences in functional and morphological brain features in healthy subject related to this polymorphisism. Most of these studies have used data from magnetic resonance imaging (MRI). However, the number of studies is small, and no one integrates multimodality neuroimaging in their analysis.
Aims
The aim of the present study is to compare the functional and structural differences in the brain of healthy subjects with BDNF Val66Met polymorphism.

Methods
Resting-state functional MRI images, structural T1 and T2 images and diffusion tensor imaging (DTI) from the UK Biobank will be analyzed using standard methods (Statistical parametric mapping 12 [SPM12] and FMRIB Software Library v5.0 [FSL] routines will be used to perform this task). The statistical analysis will consist of a comparison among the possible genotyping groups.

Project duration
6 – 9 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterization-of-genetic-and-phenotypic-of-obstructive-sleep-apnea-and-the-potential-risk-factor-of-stroke

Characterization of genetic and phenotypic of obstructive sleep apnea and the potential risk factor of stroke.

Last updated:
ID:
77803
Start date:
6 July 2022
Project status:
Current
Principal investigator:
Professor Guoqing Zhang
Lead institution:
Shanghai Institute of Nutrition and Health, CAS, China

Sleep-disordered breathing is a multiple phenotypes disorder that influence healthy status. The American Academy of Sleep Medicine (AASM) divided sleep disordered breathing into five categories, with obstructive sleep apnea (OSA) being the most common type. The most common complications of OSA are hypertension (75.9%), obesity (74.2%) and diabetes (34.1%), and it’s also a risk factor for multiple cardiovascular diseases, cognitive deficit, and stroke, the occurrence and development of OSA is closely related to complicated environment and living habits, as well as to genetic variation. Genome-Wide Association Studies (GWAS) revealed the genetic characteristics of sleep apnea, suggesting a link between sleep and genes. With the increasing prevalence of OSA, the risk of disease, disease-related phenotypes and genetic variation loci can be more detailed studied in different populations.
Exploring phenotypic features of OSA based on epidemiological data and constructing a casual-effect network of risk factors is critical. By integrating the phenotypic and genomic data of OSA population in the UK and China, we would find the phenotypic and genetic association in different population and compare the difference in both populations, identify the risk factors of OSA. Continuous Positive Airway (CPAP) Pressure is the optimal management of OSA, which can reduce cumulative incidence of stroke patients with OSA. What’s more, we observed some stroke patients also be diagnosed with OSA. CPAP for patients with OSA and stroke can treat both diseases at the same time, but CPAP requires at least 4h per night, so it is difficult to guarantee the treatment compliance. If more phenotypic associations can be found between the two diseases, it is hoped that more treatments could be developed.
We wish to study the potential association and risk factors between stroke and OSA. So as to broaden the means to promote the prevention, diagnosis, treatment and prognosis of OSA and stroke.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterization-of-genetic-modifiers-associated-with-phenotypic-variability-of-hong-kong-and-uk-spinal-muscular-atrophy-patients

Characterization of Genetic Modifiers associated with phenotypic variability of Hong Kong and UK Spinal Muscular Atrophy patients

Last updated:
ID:
199247
Start date:
18 June 2024
Project status:
Current
Principal investigator:
Mr Louis She
Lead institution:
University of Hong Kong, Hong Kong

SMA is a genetic disorder that affects the spinal cord motor neurons, leading to weakness and atrophy of muscles. It is one of the leading genetic causes of death for infants. While all cases involve a problem with the SMN1 gene, individuals can experience very different symptoms – from difficulty moving as a baby to being able to walk as an adult.
We know this difference is partially due to extra copies of a related gene called SMN2. However, even people with the same number of SMN2 copies may have varying symptoms. Scientists believe other genetic factors are also at play. Most previous research has focused on families of European descent. But genetic factors can be distinct in different communities worldwide.
In this study, we aim to better understand how genes influence SMA by comparing genetic information from two groups – individuals of British descent in the UK Biobank study, and our local cohort from Hong Kong. With approval, we will analyze half a million genetic markers across the genome. Our goal is to see which previously identified genetic factors related to SMA severity are shared between communities, and find any new ones unique to one group.
We will also look at how multiple genetic variants together might modify SMA. No single gene completely determines a person’s outcome – the combined effects of genetic variants provide a more complete picture. Comparing data internationally allows us to learn which genetic risk factors are universal, versus population-specific.
The ultimate goal is to advance personalized prediction and care for SMA patients. If we can better understand why similar children may follow different disease paths, doctors may one day tailor treatment plans based on a child’s unique genetic profile. More broadly, investigating genetic health impacts in diverse populations worldwide promotes equal opportunities for all communities to benefit from scientific progress. This project is an important step towards that objective.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterization-of-patients-with-memory-symptoms-a-longitudinal-study-of-clinical-and-brain-imaging-phenotypes

Characterization of patients with memory symptoms: a longitudinal study of clinical and brain imaging phenotypes

Last updated:
ID:
92288
Start date:
22 November 2022
Project status:
Current
Principal investigator:
Dr Veronica Raquel Alheia Cabreira
Lead institution:
University of Edinburgh, Great Britain

Our overall aim is to explore the predecessors and outcomes of memory symptoms, namely the multiple disease trajectories and distinct clinical profiles that characterize different cognitive disorders. The project aims to integrate clinical and brain imaging biomarkers, cognitive and personality traits/mood data to further define individual phenotypes of patients with non-neurodegenerative memory complaints, using follow-up data. We are interested in studying what distinguishes patients with memory complaints with and without an underlying dementia diagnosis at 5 years, and furthermore explore what characteristics (including sociodemographics and health-related behaviors) predispose to memory complaints in patients who will not develop a dementia. This will give us insight into the pathophysiology of other cognitive disorders outside the dementia scope, namely functional cognitive disorders. We will use cognitive data to investigate if certain patterns of cognitive impairment (e.g. attention/executive functions) point to a non-neurodegenerative etiology and will attempt to define what predicts a worse cognitive prognosis in these patients (comparing four groups – those with symptoms and normal cognitive performance, symptoms and impairment with reversion, impairment who remain stable and those with initial impairment that keep progressing but still do not fit into a dementia diagnosis). For those individuals with a repeat imaging visit, we intend to explore brain MRI (structural) data and look for an association between memory complaints and global brain/grey matter atrophy, in patients without a dementia diagnosis. We will use the primary care and NHS record linkage to ascertain patients with memory complaints including those referred to memory clinics. Our project meets UK Biobank’s purpose of improving the prevention, diagnosis and future treatment of cognitive illnesses. Insights from this work will lead to a better understanding of disease processes underlying cognitive decline including neurodegenerative disorders such as Alzheimer’s disease, ultimately improving the diagnosis, prognostication, and better selection for clinical trials of newly developed disease modifying-treatments. The project will make full use of phenotypic and derived brain MRI biomarker data. To maximize power, the full patient cohort with cognitive follow-up data will be used (20 346 individuals who underwent a repeat assessment five years after their initial assessment). The project provides insights to the relative contribution that social, environmental and psychological factors have on cognitive functioning. This could contribute to shape health policies to support brain and mental health. These findings will be critical for providing information to health professionals to ease clinical decision making in primary care and memory clinics.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterization-of-risk-factors-and-biomarkers-for-digestive-tract-diseases

Characterization of risk factors and biomarkers for digestive tract diseases

Last updated:
ID:
83339
Start date:
18 March 2022
Project status:
Current
Principal investigator:
Dr Hao Chen
Lead institution:
Guangdong Provincial People's Hospital, China

The digestive tract is important in maintaining the function of the human body. Diseases of the organs in the digestive tract are harmful to our health. In recent years, different methods with promising therapeutic effects have been developed to treat digestive tract diseases. Despite advances in therapeutics, digestive tract diseases can lead to serious consequences and long-lasting burdens. Thus, “the best fight is the one you avoid”, the prevention and prediction of digestive tract diseases are as important as treating them. Individual characteristics, such as lifestyles, psychological factors and genetic variations, could have far-reaching impacts on the development of digestive tract diseases. However, there are other factors that remain controversial, and indicators with high predictive sensitivity and accuracy remain deficient. It is important to provide such evidence based on large-scale studies.
In this 3-year project, we aim to 1) explore the risk factors that predispose people to the development of digestive tract disease. 2) identify indicators that can assist with the individualized prediction of digestive tract diseases. Our findings will help the public to take actions, for example, lifestyle modification, to prevent digestive tract diseases, and assist in the decision-making process of healthcare providers.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterization-of-somatic-variation-and-its-association-with-disease-states-in-the-uk-biobank

Characterization of somatic variation and its association with disease states in the UK Biobank

Last updated:
ID:
84142
Start date:
21 April 2022
Project status:
Closed
Principal investigator:
Dr Tanya M Teslovich
Lead institution:
FL2020-004, Inc., United States of America

While our genetic code is handed down to us before birth, our personal genome is dynamic and changes as we age. As our genes change over time, this creates distinct cell populations in our body – a process called somatic mosaicism. These cell mosaics bring distinct disease risks and define a new avenue of genetic susceptibility to human disease. We seek to leverage UK Biobank data to study how the genetic code that people are born with, combined with somatic mosaicism, leads to the development of age-related diseases. We aim to identify mechanisms that we can target therapeutically to prevent and treat disease. Our goal is to develop and apply these insights to improve health outcomes, including for residents of the United Kingdom.

These findings will lead to new diagnostic and therapeutic strategies to identify and mitigate risk for age-related diseases. The potential impact of this research on public health is very broad. New risk surveillance approaches and new treatments may improve outcomes across several different disease areas, including heart disease and cancers, which are leading causes of death and key drivers of healthcare costs. It is anticipated that the results of these analyses will be published and made broadly available to the community over the next 3 years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterization-of-the-effects-of-cancer-and-cardiovascular-diseases-on-the-eyes-using-interpretable-machine-learning

Characterization of the effects of cancer and cardiovascular diseases on the eyes using interpretable machine learning.

Last updated:
ID:
99518
Start date:
20 July 2023
Project status:
Current
Principal investigator:
Dr Joachim Behar
Lead institution:
Technion - Israel Institute of Technology, Israel

The eye is a sensory organ that reacts to visible light and is part of the sensory nervous system. The eye transparent cornea provides unique, non-invasive access to the cardiovascular system and the central nervous system, which may be affected by various diseases. This research aims to reveal new mechanistic insights into the effects of different diseases on the eyes and vision. For that purpose we will develop a toolbox of vasculature and neural DFI and OCT biomarkers, classical machine learning models using those biomarkers as well as deep learning models for the task of diagnosis or risk prediction, use model explainability methods to identify a digital biomarkers subset (using the classical machine learning approach) or newly discovered patterns (using the DL approach) most associated with a given endpoint. This research will reveal new relationships between patterns in the eye and a variety of diseases and consequently form a strong bridge between fundamental, clinical and engineering research. The newly discovered biomarkers and physiological insights have the potential to set the ground for development of novel screening and diagnostic procedures leveraging eye images thus impacting public health.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterize-the-burden-of-type-2-inflammation-in-dermatological-and-respiratory-diseases

Characterize the burden of type 2 inflammation in dermatological and respiratory diseases.

Last updated:
ID:
77992
Start date:
30 November 2021
Project status:
Closed
Principal investigator:
Mrs Catia Proenca
Lead institution:
Alira Health, Switzerland

Type 2 inflammation is a type of systemic allergic response that can be associated with several respiratory and/or dermatological diseases such as asthma. More than half of people with asthma suffer from an underlying type 2 inflammatory process. These patients are at increased risk of suffering asthma attacks leading to hospitalization than asthma patients without type 2 inflammation. In addition to asthma, type 2 inflammation also plays a role in other respiratory conditions such as eosinophilic esophagitis, chronic obstructive pulmonary disease, chronic rhinosinusitis and, allergic rhinitis) and dermatological diseases such as eczema. Better understanding the patient characteristics of people with type 2 inflammation and of people with dermatological or respiratory diseases and type 2 inflammation, is important to elucidate the role of type 2 inflammation in these diseases. Additionally, understanding the treatment patterns, patient pathway and resource use of people with type 2 inflammation (defined based on blood biomarkers) will be useful to understand the burden of type 2 inflammation for the health system. This study is expected to take a year to complete.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterizing-alzheimers-disease-and-dementia-in-south-korean-cohorts-by-distillating-knowledge-from-large-scale-data

Characterizing Alzheimer’s Disease and Dementia in South Korean Cohorts by Distillating Knowledge from Large-scale Data

Last updated:
ID:
96217
Start date:
6 July 2023
Project status:
Current
Principal investigator:
Professor Won Hwa Kim
Lead institution:
Pohang University of Science and Technology, Korea (South)

In this project, we will develop Artificial Intelligence (AI) frameworks to understand Alzheimer’s Disease (AD). Teaching an AI machine to investigate AD requires large amount of data. Unfortunately, existing AD datasets in South Korea are in small scale, which are not sufficient to develop AI methods. In this regards, we will use the AD data from UK Biobank to first teach an AI machine, and then apply the trained AI machine to investigate AD cohorts in South Korea. For this, we propose the following two aims:

Aim 1: We will develop an AI model to characterize AD using multi-modal data. We will use both brain images (e.g., MRI) and gene data from the UK Biobank to understand AD.

Aim2: We will develop an AI model to analyze data from multiple sites. As the data collected from different sites may have different site-specific characteristics, we will develop an AI model that neglects such effect and extract only the AD-related patterns from the data.

The frameworks developed in this project has potential to be adopted the research of other countries with insufficient data and help understanding of AD progression that stem from regional differences.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterizing-brain-changes-across-the-life-span-a-missing-link-to-cognitive-dysfunction

Characterizing brain changes across the life span – a missing link to cognitive dysfunction

Last updated:
ID:
71022
Start date:
12 April 2021
Project status:
Current
Principal investigator:
Dr Saima Hilal
Lead institution:
National University of Singapore, Singapore

Aim: It has been suggested that brain changes across the life span are strong predictors of cognitive functioning at older ages. Unfortunately, there is no data on brain damage across the life span in both Asian and European countries. We aim to find causes for cognitive dysfunction in the multi-ethnic populations from Singapore and UK and identify individuals who might be at high risk of cognitive decline and other clinical events. We will study early-aged, middle-aged, and late-life individuals by using the information on their lifestyle, blood tests, and brain MRI scan. We will then link these data with information on cognitive function. We will also study how changes in lifestyle, brain, and blood markers affect cognition over time and increase the risk of cognitive decline.

Scientific rationale: we will be able to identify factors and processes that determine why some people develop cognitive impairment and others do not develop this disease. This knowledge will inform the development of strategies for health promotion, and the design of personalized interventions tailored to the individual’s specific behavioral risks such as lifestyle modification.

Project duration: 3 year period

Public health impact: This research will identify new opportunities to improve public health policies, resulting in better interventions and management of cognitive decline at an early stage. This will enable clinicians to make earlier diagnoses, identify those at risk, and prevent the development of cognitive decline according to the risk profile in precision aging public health.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterizing-brain-networks-and-their-inter-individual-variability-by-high-throughput-imaging-and-computational-modelling

Characterizing brain networks and their inter-individual variability by high-throughput imaging and computational modelling

Last updated:
ID:
41655
Start date:
26 April 2019
Project status:
Current
Principal investigator:
Professor Simon Eickhoff
Lead institution:
Research Center Juelich, Germany

The human brain shows a marked level of neurobiological variability between individuals. Likewise, individuals also vary widely in aspects that may be considered causes (age, gender, general health, environmental factors) and effects (cognitive functions, socio-affective traits) of this neurobiological variability. However, the relationship between neurobiological and behavioural variability relate to each other, is still enigmatic. The goal of this project is to probe the relationship between brain features on one hand and a broad range of behaviours on the other hand in order to provide an overview on brain-behaviour and brain-environment relationships.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterizing-malignant-hyperthermia-prevalence-and-risk-factors-within-a-large-uk-general-population-cohort

Characterizing Malignant Hyperthermia prevalence and risk factors within a large UK general population cohort.

Last updated:
ID:
290317
Start date:
30 January 2025
Project status:
Current
Principal investigator:
Dr Vivianne Tawfik
Lead institution:
Stanford University, United States of America

Malignant hyperthermia (MH) is a muscle genetic disorder that is triggered by common anesthetic agents. Although rare, if not recognized early, MH can lead to serious complications and possibly death while undergoing an operation. Our understanding of MH has improved over the years with the adoption of genetic sequencing, however, more work is needed to further understand which patients are at the highest risk of an MH crisis. Given the ubiquity of anesthesia worldwide and the consequences of not recognizing MH early, further understanding of MH and its risk factors has the potential to save lives. The study should take approximately 1-2 years to complete.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterizing-penetrance-and-phenotypic-spectrum-of-disease-in-general-population-cohorts

Characterizing penetrance and phenotypic spectrum of disease in general population cohorts

Last updated:
ID:
922592
Start date:
30 October 2025
Project status:
Current
Principal investigator:
Dr Brittany Sears
Lead institution:
Ambry Genetics Corporation, United States of America

Gene-disease relationships are typically established in individuals or families with a disease burden markedly higher than the general population. This sampling bias can potentially create bidirectional error in estimating the risk of health conditions associated with genetic differences by 1) inflating estimation of risk due to over-representation of high-penetrance families, and 2) limiting described spectrum of disease described for variants when phenotype is used for ascertainment. In many syndromes with germline genetic causes, accurately characterizing spectrum of disease, lifetime risk, and age of onset is essential to providing recommendations for clinical intervention. We propose to use the prospective UK Biobank cohort for two research aims: 1) comparing cancer penetrance and age of onset in individuals with germline pathogenic variants in hereditary cancer risk genes in this cohort versus our retrospective patient data, and 2) characterizing the phenotypic spectrum and penetrance in additional rare diseases, such as neurodevelopmental disorders. We will utilize whole genome sequencing available in the database along with phenotype (e.g., ICD10 codes, cancer site), genetic sex, and relatedness to other individuals in the Biobank to characterize spectrum of disease in this prospective cohort that is likely to be less affected by ascertainment bias than in commercially tested patient populations where from which research has historically drawn. This novel approach to characterizing penetrance and spectra of disease in the general population serves the public good by better informing tailored risk management and anticipatory guidance for individuals with germline genetic predispositions to disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterizing-the-association-and-causation-between-novel-open-reading-frames-and-disease

Characterizing the association and causation between novel open reading frames and disease.

Last updated:
ID:
109483
Start date:
31 January 2024
Project status:
Current
Principal investigator:
Professor Yi-Hsiang Hsu
Lead institution:
ProFound Therapeutics, Inc., United States of America

To understand how specific changes in a person’s DNA can cause disease, scientists rely on a “map” of annotations that describes which portions of the genome contain instructions for making which proteins. The standard map scientists use for this purpose has only 2% of genomic regions filled in with protein-coding annotations. However, many additional regions have the potential to code for proteins.
For this project, we plan to compare a map of new protein locations with the genetic information of participants in the UKBiobank Project. Our aims are to:
1. Identify groups of healthy individuals those affected by similar diseases that can be compared to each other.
2. Determine which novel proteins are changed or missing in both groups of participants.
3. Use statistics to determine which of these novel proteins are likely connected to each disease.
Finding these novel proteins that are associated with disease will allow scientists to identify new ways to develop therapies that might help people suffering from those diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterizing-the-contribution-of-short-tandem-repeats-to-human-phenotypes

Characterizing the contribution of short tandem repeats to human phenotypes.

Last updated:
ID:
46122
Start date:
10 April 2019
Project status:
Current
Principal investigator:
Dr Melissa Gymrek
Lead institution:
University of California, San Diego, United States of America

Short Tandem Repeats (STRs) are a class of genetic variation comprising of repeated short sequences of DNA in the genome. Several dozen STRs are known to contribute to human diseases, including Huntington’s Disease and Fragile X Syndrome. However there are more than 1 million STRs in the human genome, most of which remain uncharacterized.

Traditionally studying the role of STRs has been difficult since they are complex to analyze and are not directly captured by most genetics studies. We recently developed a resource that allows to analyze STRs in large datasets where they were not directly genotyped using a technique known as imputation. Here, we will leverage this resource to identify the contribution of STRs to a variety of traits in humans.

We expect that imputing STR in the UK Biobank data and performing association tests will take up to 1 year with 1-2 years follow up work to evaluate and interpret our results. We expect our study will identify a novel class of genetic variation with widespread impact on a variety of human traits.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterizing-the-determinants-and-consequences-of-clonal-hematopoiesis

Characterizing the determinants and consequences of clonal hematopoiesis

Last updated:
ID:
186376
Start date:
22 October 2024
Project status:
Current
Principal investigator:
Dr Abhishek Niroula
Lead institution:
University of Gothenburg, Sweden

Clonal hematopoiesis (CH) is defined by the presence of shared acquired genetic mutations in a large fraction of blood cells. CH is a common phenomenon in the elderly. Presence of CH is associated with a higher risk of blood cancer, mortality, and a wide range of diseases, including cardiovascular, pulmonary, and liver diseases.
We and others have identified genes that are recurrently mutated in CH. These genes are also commonly mutated in blood cancer. Recent studies have revealed that the majority of CH do not harbor mutations in previously known genes. Moreover, the mechanisms of how CH develops and leads to diseases are not understood.
This project aims to identify new genetic alterations in CH and discover mechanisms influencing CH and the pathogenesis of CH-mediated diseases. The massive sample size and coverage of the sequencing data in UK Biobank provides opportunities to identify new genetic variations and regulators of CH and their implications on human health. We will utilize sequencing data from approximately half a million participants in the UK Biobank to
1) characterize genetic mutations involved in CH,
2) identify genetic and environmental factors that influence the risk of developing CH, and
3) develop methods to predict risks associated with CH mutations.
This project will identify several new genetic alterations in CH and reveal novel mechanisms of CH development. These findings may provide opportunities for a) stratifying risk, b) early diagnosis, and c) develop new therapies for treating CH-mediated diseases. The genetic alterations identified here may serve as markers for screening of CH in the longer run. The findings will be shared with other researchers and published in scientific journals and meetings.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterizing-the-genetic-and-biological-heterogeneity-of-alzheimers-disease-risk

Characterizing the genetic and biological heterogeneity of Alzheimer’s disease risk

Last updated:
ID:
63648
Start date:
12 January 2021
Project status:
Closed
Principal investigator:
Dr Jeremy Elman
Lead institution:
University of California, San Diego, United States of America

Alzheimer’s disease (AD) is often thought of as a single disease with a standard set of features. However, research suggests there is much variability across people. For example, prior studies find individuals can have different types of cognitive impairment (“cognitive subtypes”). People can also show different patterns of tissue loss or pathology across the brain (“biological subtypes”). Interestingly, the different patterns of brain changes are associated with different cognitive impairments. This suggests that even “typical” AD may represent multiple disease subtypes. It is unclear whether these subtypes have different causes that require different treatments. Examining genetic risk can help us answer this question, yet hundreds or thousands of genes may be involved in a disease like AD. One approach is to sum all of these effects into a single value for each person, known as a polygenic risk score (PRS). However, this only considers risk along a single continuum: high versus low. Two individuals might have the same risk score, but very different patterns of which genes are contributing to this risk. These differences may cause different forms of the disease. This project will seek to identify genetic subtypes of AD by creating multiple PRSs for each person that summarize risk in sets of genes with related effects, often referred to as biological pathways. This project will address two key questions: 1) Does the genetic risk for AD exist along multiple dimensions, not just high versus low; and 2) how does variability in the genetic risk for AD relate to the differences in the disease we see at the level of brain and cognition? The specific aims are to: 1) determine whether there are subtypes of AD genetic risk using PRSs specific to biological pathways; 2) test whether different patterns of brain tissue loss and pathology are associated with different forms of AD genetic risk; and 3) examine whether differences in cognitive impairment are associated with different forms of AD genetic risk. The project will use UK Biobank data to identify genetic and biological subtypes of AD that will be used to test for relationships in independent. The project duration will be ongoing, with an initial period of 36 months. Understanding the genes and biological pathways that may cause different types of AD will improve efforts to determine which types of treatment are likely to be most effective for each AD subtype.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterizing-the-genetic-and-environmental-underpinnings-of-psychiatric-disease-and-associated-phenotypes

Characterizing the genetic and environmental underpinnings of psychiatric disease and associated phenotypes

Last updated:
ID:
84157
Start date:
28 October 2024
Project status:
Current
Principal investigator:
Dr Keira Johnston
Lead institution:
Yale University, United States of America

Psychiatric disorders, such as major depression, eating disorders and post-traumatic stress disorder (PTSD), are often difficult to treat & the way they develop is not fully understood. Chronic pain, pain that lasts 3 months or longer, is a common experience and is associated with a wide range of different conditions, including psychiatric disorders, or can be thought of as a condition in and of itself. The reasons why some individuals’ chronic pain develops whereas others remain pain-free, and the mechanisms behind this, are not fully understood. Chronic pain can also be difficult to effectively treat. When chronic pain and psychiatric disorder are experienced together, this can have a negative impact on the chances of effective treatment of either condition.

Eating disorders occur in people of all sexes and genders and in people of all ages. Currently, the most well studied eating disorder is anorexia nervosa (AN) which is most common in adolescent females. Researchers gathered cases of patients with AN and patients without, and learned that there are genetic factors as well as environmental factors that contribute to AN risk. We learned that the genetic risk factors of AN also play a role in other psychiatric disorders like obsessive compulsive disorder and metabolic factors like insulin resistance and other hormonal signaling. We still have a lot to learn about what the metabolic risk factors are and how they act in the body to promote disordered eating and how we can treat that inherited risk. We also have a lot to learn about how eating disorders present in males, older individuals, and those that do not meet criteria for Anorexia Nervosa but have some features of the condition or who do not have restrictive eating behaviors and instead have binge eating behaviors.

Previous genetic investigations have shown sections of DNA associated with chronic pain and with different psychiatric disorders, but the exact way in which these small changes in DNA result in the development of chronic pain and of psychiatric disorder is not fully understood. Studies trying to investigate mechanisms of development such as brain imaging studies, are often expensive to carry out and have limited sample size. The approaches we will take allow for very large, well-powered study into the causal contributing factors in the development of chronic pain and of psychiatric disorder, potentially improving treatment options and understanding of these conditions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterizing-the-genetic-determinants-of-mineral-metabolism-a-uk-biobank-study

Characterizing the genetic determinants of mineral metabolism: A UK Biobank study

Last updated:
ID:
45742
Start date:
7 May 2019
Project status:
Closed
Principal investigator:
Fadil Hannan
Lead institution:
University of Oxford, Great Britain

Aims: The purpose of this study is to identify common variations within the human genome, which influence the concentration of minerals in the blood such as calcium and phosphate. Alterations in these minerals are also linked to major diseases such as osteoporosis and cardiovascular disease, and this study will investigate whether genetic variations that regulate blood minerals also contribute to the occurrence or severity of these major diseases.

Scientific rationale: Calcium and phosphate are required to mineralise bone, and this process is regulated by alkaline phosphatase, which is a protein produced by bone cells. Disturbances in the blood concentrations of calcium, phosphate and alkaline phosphatase have been linked to osteoporosis, which is a common disorder characterised by reduced bone strength and an increased susceptibility to broken bones. Alterations in blood calcium and phosphate also predispose to cardiovascular diseases such as heart attacks and angina. The circulating concentrations of calcium, phosphate and alkaline phosphatase in healthy individuals are considered to be influenced by multiple different variations within the human genome, however, many of these genetic variations remain to be identified. We will undertake a genome-wide association study, which is a technique used to rapidly scan the entire genome for variations that occur commonly (in >1% of the population). We will then evaluate whether these genetic variations are linked to calcium, phosphate and alkaline phosphatase concentrations in blood samples collected from UK Biobank participants. Genetic variations that are found to be linked to these blood mineral parameters will be further evaluated for associations with osteoporosis and cardiovascular diseases.

Project duration: 24 months

Public health impact: Increased understanding of the genetic factors that control the blood concentrations of calcium, phosphate and alkaline phosphatase may lead to the discovery of new drug targets for disorders of bone and mineral metabolism, and also for cardiovascular diseases. Furthermore, this study has the potential to advance personalised medicine by identifying individuals that may be susceptible to side effects from drugs that influence mineral metabolism such as calcium supplements, vitamin D preparations, and osteoporosis drugs.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterizing-the-impact-of-shocks-in-clinical-care-on-disease-onset-prediction-and-prognostic-algorithms-a-focus-on-the-covid-19-pandemic

Characterizing the impact of shocks in clinical care on disease onset, prediction, and prognostic algorithms: a focus on the COVID-19 pandemic

Last updated:
ID:
76125
Start date:
13 May 2022
Project status:
Current
Principal investigator:
Dr Scott Logan Lipnick
Lead institution:
Flagship Pioneering Labs TPC, INC, United States of America

The proposed work will focus on establishing systems to identify individuals at varying state changes in their health and exploring whether these models can be deployed across time (i.e., pre-Coronavirus Disease 2019 (COVID-19) through the current era). Importantly, our assumption – based on multiple studies with quality evidence – is that explainable models will be able to be developed from longitudinal data that can be used for present-day interventions. Given the power of the UK Biobank data, we intend for our work to enable disease or dysfunction detection before major changes in health. Our aims are to:
1. Establish cohorts of participants likely affected by similar biological and clinical diseases
2. Characterize their health-to-disease progression
3. Build predictive platforms to identify these individuals before major state changes
4. Interrogate how portable these predictive platforms across eras with different medical utilization such as before, during, and after the COVID-19 pandemic.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterizing-the-incidence-of-acmg-reportable-secondary-findings-and-clinical-outcomes-in-a-large-prospective-cohort

Characterizing the Incidence of ACMG Reportable Secondary Findings and Clinical Outcomes in a Large Prospective Cohort

Last updated:
ID:
60148
Start date:
8 December 2021
Project status:
Closed
Principal investigator:
Dr Zachary Laksman
Lead institution:
University of British Columbia, Canada

Many rare cardiac diseases can be diagnosed through analysis of the patient’s genome. Modern techniques analyze a patient’s entire set of genes, or their genome. Although effective, these tests can result in incidental/secondary findings, where the test reveals genetic mutations unrelated to the original reason for testing. Mutations to some genes can result in potentially lethal diseases, and therefore the American College of Medical Genetics and Genomics created recommendations for diagnostic laboratories and clinicians to disclose these secondary findings to patients. These recommendations include several cardiac-related genes. Consequently, patients must be seen by medical specialists and be subject to medical testing and possibly interventions. However, many individuals carry genetic mutations and the presence of a mutation does not always confer disease. This leaves many asymptomatic patients with unnecessary medical attention and increased anxiety. Furthermore, these patients may potentiate wait times for specialists and increase healthcare spending.

The aim of our research is to study the burden of secondary findings related to heart diseases from genomic testing on the population-level. We will use the public genetic mutation database ClinVar to identify genetic mutations associated with actionable genetic secondary findings. We will then quantify the incidence of secondary findings in the UK Biobank, a large prospective cohort containing genomic and clinical data from approximately 500 000 participants. Subsequently, we will analyze clinical data for the UK Biobank participants carrying these reportable genetic variants and classify participants with a positive or negative disease phenotype, allowing us to analyze the relationship between genotype and clinical outcomes within the context of the American College of Medical Genetics and Genomics reportable secondary findings list. We hypothesize that there will be a significant number of people within the UK biobank that have reportable cardiac genetic findings per current guidelines, however many will not have clinical findings related to a disease.

We anticipate this project to be completed 12 months after gaining access to the required data. This research will describe the scope of the genetic secondary findings issue and inform future system-level policies and interventions which will improve the quality of patient care and reduce expenditures. This research will also provide a foundation for future projects which may further describe the economic costs of secondary findings. Ultimately, we hope to decrease the number of patients who are unnecessarily being given medical attention due to their harmless genetic mutations.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterizing-the-inter-relationships-between-polypharmacy-and-multiple-long-term-conditions-in-uk-biobank

Characterizing the inter-relationships between polypharmacy and multiple long-term conditions in UK Biobank

Last updated:
ID:
69836
Start date:
1 March 2021
Project status:
Current
Principal investigator:
Professor Nick John Reynolds
Lead institution:
Newcastle University, Great Britain

People who live with a number of medical conditions (multiple long-term conditions or MLTCs) are at high risk of poor health. They are often prescribed multiple medicines. When the number of medicines is greater than five, this is called polypharmacy. The relationship between MLTCs and polypharmacy is complex and not well understood. We know that some patients enter a downward spiral, developing an increasing number of conditions and being prescribed more and more medicines. This can cause health problems, as individual medicines may interact with one another or have side-effects. Other medicines may modify the downward spiral by preventing the development of conditions such as heart disease and cancer. All of this makes it difficult to design interventions to ensure medicines are prescribed in combinations that do more good than harm.
Our long-term goal is to better understand the dynamic relationship between MLTCs and polypharmacy, to optimise the medicines prescribed for individual patients. This research will also identify key points for intervention, to maintain the best possible health trajectory for people with MLTCs.
Our group has experience in applying new developments in computer technology, termed artificial intelligence (AI) and machine learning, to healthcare data. We will develop these methods to look for new patterns linking MLTCs and prescribed medicines within UK biobank data. The information and patterns generated will feed into the design of a larger collaborative project accessing electronic care records in the North East of England and East London.
PPI helped to shape the proposed research, from the title and questions being addressed, through to the dissemination strategy. Because of the complexity of MLTCs, the many risk factors for them, and the sensitivity of data in electronic healthcare records, PPI is embedded across all aspects of our work.
In the long-term, our research will lead to an alert system in medical records and strategies for prevention and improved management of multiple long-term conditions. With PPI, we will find the best way of sharing findings from our work with diverse audiences. This will include insights into effective ways of forming multidisciplinary research teams and PPI for MLTCs. As a minimum, we will use webinars and social media, conferences presentations and scientific publications.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterizing-the-multimodal-determinants-of-therapy-related-cardiac-dysfunction-in-breast-cancer-survivors-a-uk-biobank-based-study

Characterizing the multimodal determinants of therapy-related cardiac dysfunction in breast cancer survivors: A UK Biobank-based study

Last updated:
ID:
1006098
Start date:
24 September 2025
Project status:
Current
Principal investigator:
Dr Fan Yang
Lead institution:
Sichuan Cancer Hospital & Institute, China

Scientific Rationale:Thanks to therapeutic advances, breast cancer survival rates have significantly improved, but cardiovascular disease (CVD) has become a leading cause of morbidity and mortality among survivors, with risks comparable to cancer recurrence. Key treatments such as anthracyclines, targeted therapies, and radiotherapy all carry cardiotoxic risks. However, our understanding of the true incidence of various CVD subtypes, effective early predictive biomarkers, and genetic susceptibility remains limited. The UK Biobank, with its prospective cohort of over 500,000 individuals, long-term follow-up records, and deep multimodal data (cardiac imaging, proteomics, genomics), offers a unique platform to systematically address these critical questions in cardio-oncology.
Research Questions & Objectives:
This study aims to leverage the UK Biobank’s multimodal data to comprehensively characterize the determinants of cancer therapy-related cardiac dysfunction (CTRCD).
Core Questions:
1) What is the long-term cardiovascular risk for breast cancer survivors?
2) Can new imaging and proteomic biomarkers be identified for early prediction?
3) Is there a genetic predisposition to cardiotoxicity?
Specific Objectives:
Quantify Risk: To precisely calculate and compare the incidence rates of various CVDs (e.g., heart failure, myocardial infarction) between breast cancer survivors and matched controls, and to assess the synergistic effect of a cancer diagnosis with baseline risk factors.
Discover Biomarkers: To integrate cardiac magnetic resonance (CMR) imaging phenotypes and circulating protein profiles to identify novel, non-invasive biomarkers that predict subclinical cardiac injury and long-term CVD risk.
Elucidate Genetic Mechanisms: To identify genetic variants associated with CTRCD through genome-wide association studies (GWAS) and to develop a polygenic risk score (PRS) for more accurate, individualized risk stratification.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterizing-the-phenotypic-spectrum-of-sickle-cell-disease-patients-and-individuals-with-sickle-cell-trait-using-deep-phenotyping-and-genomic-data

Characterizing the phenotypic spectrum of sickle cell disease patients and individuals with sickle cell trait using deep phenotyping and genomic data

Last updated:
ID:
78920
Start date:
8 November 2021
Project status:
Closed
Principal investigator:
Dr Onuralp Soylemez
Lead institution:
Global Blood Therapeutics, Inc, United States of America

Scientific rationale: this study aims to further our understanding of the genetic basis of variation in blood traits between individuals. While some blood traits such as hemoglobin levels or red blood cell counts are routinely monitored for overall health status, variation in these traits may also help us understand the biological underpinnings of many inherited blood disorders such as sickle cell disease (SCD). SCD is a common inherited blood disorder that disproportionately affects individuals of African ancestries. There is significant variation in clinical presentation and treatment response of patients with SCD. Due to a paucity of medical and genetic data in underserved communities, our understanding of the full spectrum of the medical symptoms and lab measurements in SCD patients and those at risk for SCD related medical complications is incomplete. We aim to study the impact of genetic variation in blood traits on health outcomes in individuals with SCD and those without SCD.
Aims: we will catalogue the full extent of medical symptoms in SCD patients in the UK Biobank cohort using extensive electronic medical records, blood measurements from different time points, lifestyle and genetic data. We will investigate the impact of genetic differences between individuals on differences in their health outcomes including SCD or its common complications such as kidney failure, lung problems and stroke. This will help us identify lab measurements that can predict the disease severity or progression to inform patient management.
Project duration: this study aims to heavily utilize the DNA sequencing data linked to long-term health outcomes and lab measurements, which will be released with rolling updates. Therefore, we estimate the project duration to be three years. We are fully committed to sharing our research findings with the scientific community and making our results publicly available in accordance with the annual update requirement.
Public health impact: SCD is a devastating blood disorder characterized by chronic severe anemia that can lead to a wide spectrum of end organ damages. There are approximately 15,000 people in the UK with SCD, more than 350,000 babies are born each year all over the world with SCD. These patients suffer ~30-year reduction in life expectancy due to SCD and its complications. SCD presents an urgent unmet need in underserved communities. We believe the large-scale UK Biobank data will provide genetic insights into blood traits resulting in better understanding of red blood cell health and treatment opportunities for these patients.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterizing-the-proteome-of-patients-with-polymyalgia-rheumatica

Characterizing the proteome of patients with polymyalgia rheumatica

Last updated:
ID:
751650
Start date:
31 July 2025
Project status:
Current
Principal investigator:
Dr Claire Elizabeth Owen
Lead institution:
University of Melbourne, Australia

Background: Polymyalgia rheumatica (PMR) is a highly prevalent immune-mediated rheumatic disease that results from inflammation of the musculotendinous structures surrounding the shoulders, hips and knees. Clinically, this manifests with severe pain and stiffness at affected joints and muscles. This description of PMR has only become possible courtesy of modern imaging technology including 18F-fluorodeoxyglucose positron emission tomography (18F-FDG PET/CT) and magnetic resonance imaging (MRI). A highly sensitive and specific biomarker to aid diagnosis or measure disease activity has never been discovered in PMR. Moreover, treatment of this condition still comprises prednisolone monotherapy in the first instance, the harms of which are now well recognised. Better diagnostic tools and novel therapeutic approaches represent key unmet needs in the management of patients with PMR.

Hypothesis: That the peripheral blood of patients with PMR exhibits a unique proteomic signature that distinguishes it from normal controls and individuals with other closely related or analogous autoimmune conditions.

Objectives:
1) To profile the plasma proteome of patients in the UK Biobank with a diagnosis of PMR, specifically utilising existing Olink Explore 3072 data.
2) To contrast the “PMR proteome” with that of normal controls.
3) To determine similarities and differences in the plasma proteome of PMR patients and the overlapping large vessel vasculitis, giant cell arteritis ([GCA], also known as temporal arteritis), along with the life-threatening complication of aortic aneurysm.
4) To explore relationships that exist between the proteomic signature of PMR and other common autoimmune conditions including rheumatoid arthritis (RA), related entities of the spondyloarthropathies (psoriasis, ulcerative colitis) and systemic lupus erythematosus (SLE).


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterizing-the-relationship-between-brain-and-genetic-cognition-and-environmental-in-ad-continuum

Characterizing the relationship between brain and genetic, cognition and environmental in AD continuum

Last updated:
ID:
176809
Start date:
17 April 2025
Project status:
Current
Principal investigator:
Ms Huijuan Chen
Lead institution:
Hainan Medical University, China

Alzheimer’s Disease (AD) affects millions worldwide, with cases expected to rise from 36 million today to over 100 million by 2050. Given the limited treatment options, early detection and intervention are crucial. Our project utilizes the UK Biobank’s extensive datasets, including brain images and genetic data, combined with health assessments such as cognitive scores and lifestyle factors, to uncover early signs of Alzheimer’s and factors influencing its progression. Over the next three years, we will use advanced statistical and machine learning techniques to: 1. Identify individuals at high risk of developing Alzheimer’s. 2. Investigate how changes in brain structure and function correlate with cognitive decline. Create predictive models for Alzheimer’s progression and risk assessment. Our research aims to enhance early detection strategies and deepen understanding of Alzheimer’s mechanisms, paving the way for more effective treatments in the early stages of the disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterizing-the-role-of-adipokines-and-additional-circulating-proteins-across-metabolically-challenged-individuals-in-predicting-end-organ-fibroinflammation

Characterizing the role of Adipokines and additional circulating proteins across metabolically challenged individuals in predicting end organ fibroinflammation

Last updated:
ID:
1015354
Start date:
26 September 2025
Project status:
Current
Principal investigator:
Dr Laurent Audoly
Lead institution:
Privebio, Inc., United States of America

Scientific Rationale
Metabolic dysfunction drives fibroinflammatory damage across multiple organs-HFpEF (heart), MASH (liver), and CKD (kidney). Adipose dysfunction releases pro-inflammatory adipokines (leptin, resistin) while reducing adiponectin. Endotrophin (ETP), a collagen VI-derived peptide, links adipose expansion to multi-organ fibrosis and represents a key therapeutic target. Identifying ETP-elevated patients using clinical variables could enable precision medicine approaches.

Research Questions:
What adipokine/proteomic signatures (especially ETP) characterize metabolic phenotypes across HFpEF, MASH, and CKD?
How do these correlate with organ-specific fibroinflammation (cardiac/liver/kidney imaging)?
Can simple clinical criteria identify high-ETP patients with multi-organ risk?

Objectives
Obj1: Analyze ETP, adipokines, inflammatory proteins in UK Biobank (n!50,000) stratified by HFpEF (NT-proBNP, diastolic dysfunction), MASH (FIB-4, steatosis), CKD (eGFR<60, albuminuria)
Obj2: Correlate proteomics with imaging: cardiac T1/ECV (fibrosis), liver MRI-PDFF/elastography, kidney morphology
Obj3: Iteratively test clinical criteria:
BMI!30 alone vs BMI+comorbidities
Identify high-ETP subtypes with multi-organ involvement
Optimize criteria for therapy selection
Obj4: Estimate prevalence of ETP-elevated phenotypes

Methodology
Stratify participants by metabolic criteria (BMI!BMI+diabetes/dyslipidemia!clinical obesity). Integrate Olink proteomics with multi-organ imaging. Use machine learning to identify ETP-associated pathways. Validate findings through cross-validation.

Expected Impact
Establish ETP as pan-organ fibroinflammatory biomarker, enable clinical identification of therapy-responsive patients, quantify multi-organ disease burden, and guide development of ETP-targeted interventions for metabolic complications.
!


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterizing-the-shared-genetic-basis-of-common-cancers

Characterizing the Shared Genetic Basis of Common Cancers

Last updated:
ID:
14105
Start date:
20 July 2015
Project status:
Current
Principal investigator:
Professor John Witte
Lead institution:
University of California, San Francisco, United States of America

The goal of our proposed research is to characterize pleiotropic loci in order to gain new insight into common carcinogenic mechanisms, shared etiology of multiple cancers, and treatment for cancer patients with seemingly distinct diseases. The UK cohort currently holds 80K subjects across multiple malignant cancer types. We propose to comprehensively assess the shared genetic basis underlying these different cancers within the UK cohort through the following aims: 1) Assess the co-inheritance of cancers due to common variation. 2) Evaluate locus-specific pleiotropy across different cancers. The detection and characterization of pleiotropy is key to understanding the biological and clinical underpinnings of cancer. While any single pleiotropic variant may have modest impact on disease, combinations of multiple variants can provide increasingly accurate prediction and be important for individualized risk counseling as well as for cancer screening and surveillance. Even where there already exist clinically relevant findings for individual cancers, our efforts to detect pleiotropy may provide an avenue for informing the successful development and application of treatments across cancers. Genotyping of all cancer subjects on the Affymetrix array is close to completion for the UK biobank subjects, and data on per subject cancer type is already available. We will use these data to determine genome-wide heritability for the most common cancers in the UK cohort, and calculate the co-inheritance and overall shared genomic basis among these cancers using complex statistical analysis methods. We request the full genotyped cohort.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/characterizing-treatment-response-in-major-depressive-disorder-using-neuroimaging-and-ehr-data-from-uk-biobank

Characterizing Treatment Response in Major Depressive Disorder Using Neuroimaging and EHR Data from UK Biobank

Last updated:
ID:
941072
Start date:
24 October 2025
Project status:
Current
Principal investigator:
Dr Anwar Said
Lead institution:
Vanderbilt University, United States of America

This study aims to investigate how multimodal data can be used to predict and understand different antidepressant medication/treatment response in individuals with Major Depressive Disorder (MDD), leveraging UK Biobank’s extensive resources.

Research questions:

1. To what extent can brain imaging data (functional MRI), demographic information and electronic health records be used with machine learning approaches to predict antidepressant treatment response in individuals with MDD?
2. What are the most effective strategies for representing and integrating heterogeneous data sources to develop robust machine learning pipelines for treatment response prediction?
3. How does stratification of the patient cohort based on clinical, demographic, or diagnoses information influence predictive model performance and the identification of clinically meaningful markers of treatment response?

Objectives:
a) To quantify antidepressant treatment response in individuals with MDD
b) To investigate whether fMRI and other scales are associated with baseline symptom severity and treatment outcomes.

Scientific rationale:
MDD is a leading global cause of disability, yet treatment efficacy is highly variable in real-world settings. Clinical trials often lack generalisability to broader populations, underscoring the need for large-scale, naturalistic evidence on treatment outcomes and predictors of response. UK Biobank provides a unique opportunity to address this gap by combining self-reported mental health data, prescription records, hospital and primary care data, demographic information, and brain imaging-derived phenotypes. This research will help identify factors associated with treatment response and inform the development of personalised treatment strategies, with the ultimate goal of improving outcomes for individuals with MDD.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/charting-the-comprehensive-landscape-of-infection-and-disease-association-using-large-scale-uk-biobank-cohort

Charting the comprehensive landscape of infection and disease association using large-scale UK biobank cohort

Last updated:
ID:
95413
Start date:
19 July 2023
Project status:
Current
Principal investigator:
Professor Joo Sang Lee
Lead institution:
Sungkyunkwan University, Korea (South)

There are cases where certain bacteria or viral infection is associated with future incidence of specific diseases. Most of these studies are focused on particular infection-disease pairs and systematic analysis on the infection-disease association landscape was not feasible due to the lack of such large-scale comprehensive data source. We believe UK biobank provides a unique opportunity to address this question to perform a systematic analysis to chart a comprehensive map of infection-disease association at a large-scale. We plan to analyze the large-scale multi-modal data provided by UK Biobank to discover the evidence for the association between the pathogen infection and the disease incidence. We will make sure to control for potential confounders such as genetic susceptibility from whole-exome (or whole-genome) sequencing data and additional clinical/physiological covariates. In addition, we might be able to find such infections that reduce (instead of increase) the incidence of specific diseases. In sum, charting the infection-disease landscape will help us developing improved disease therapeutics and preventions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/childbirth-and-depression

Childbirth and depression

Last updated:
ID:
69177
Start date:
16 March 2021
Project status:
Closed
Principal investigator:
Miss Therese Marie Johansson
Lead institution:
Uppsala University, Sweden

Postnatal depression is one of the most common psychiatric disorders, affecting more than one in every ten women within a year of giving birth. Family history of psychiatric disorders is one of the strongest risk factors for postnatal depression, suggesting that the development of postnatal depression is heritable. However, the genetic contribution to postnatal depression is to a large extent unknown.

Our overall aim with this project is to investigate how genetics contributes to the risk for postnatal depression. First, we aim to identify specific genes that might influence the risk for postnatal depression. Second, we will evaluate the genetic overlap between postnatal depression and other major psychiatric disorders. Last, we aim to elucidate whether first onset and recurrent depression in the postnatal period reflect different disorders.

The results of this study will be of great relevance to society, and especially to women in reproductive age, as our results will broaden the knowledge of how child birth affects the mental health of women. If we can identify specific genes responsible for the increased risk for postnatal depression and their biological mechanism, we could improve therapy among affected women. By investigating the possibility of estimating a woman’s genetic risk for postnatal depression, as well as the familial risk for postnatal depression, we will evaluate the use of genetic information to identify and prevent postnatal depression in high-risk women.

The data analyses will take place during 2021 and writing of manuscripts and presentation results during 2022. We expect the results from the project to be published in late 2022.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/childhood-onset-asthma-and-cardiac-structure-function-and-disease-risk-evidence-from-the-uk-biobank

Childhood-onset asthma and cardiac structure, function, and disease risk: evidence from the UK Biobank

Last updated:
ID:
198259
Start date:
5 April 2024
Project status:
Current
Principal investigator:
Professor Hu Chenkai
Lead institution:
The Second Affiliated Hospital of Nanchang University, China

Aims:
Our research aims to understand how asthma that starts in childhood may affect the heart as people grow older. We want to know if there’s a connection between childhood asthma and changes in the heart’s structure, how well it works, and if it increases the risk of heart problems later in life.

Scientific Rationale:
While we know a lot about how asthma affects breathing, we’re still learning about its impact on other parts of the body. Some studies suggest that asthma might have long-term effects on the heart, but we need to investigate this further. By studying a large group of people, we hope to find out if childhood asthma is linked to changes in the heart and if so, why that might happen.

Project Duration:
This research will take a few years (could be 2-3 years) to complete. We’ll analyse data from a group of people who have shared their health information with us over time. This long-term data allows us to see how things change and develop as people age.

Public Health Impact:
Understanding the connection between childhood asthma and heart health is important for everyone’s well-being. If we discover that there’s a link, doctors can use this information to better care for people with a history of childhood asthma. It could also help us create strategies to prevent heart problems in these individuals. Our findings might influence how doctors and policymakers approach healthcare, ensuring a more comprehensive understanding of the impact of asthma on overall health. Ultimately, our goal is to improve the health and quality of life for people who experienced asthma in their early years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/chronic-disease-outcomes-across-the-glycaemic-spectrum-harnessing-uk-biobank-and-linked-ehealth-data-to-quantify-risks-explore-mechanisms-and-determine-treatment-impacts

Chronic disease outcomes across the glycaemic spectrum: harnessing UK Biobank and linked eHealth data to quantify risks, explore mechanisms and determine treatment impacts.

Last updated:
ID:
7661
Start date:
1 January 2015
Project status:
Closed
Principal investigator:
Professor Nishi Chaturvedi
Lead institution:
University College London, Great Britain

Type 2 diabetes is associated with increases in cardiovascular disease (myocardial infarction, stroke and heart failure), cognitive decline and cancer. HbA1c, a blood test which indicates blood glucose levels (glycaemia) over the preceding 3 months, regardless of diabetes status, is available on all Biobank participants at baseline. We will examine risks of cardiovascular disease, cognitive decline and cancer across the glycaemic spectrum, including in people without diabetes. We will investigate how risks differ by gender and ethnicity. Additionally, we will explore the impact of anti-diabetic, anti-hypertensive and lipid-lowering medication on cardiovascular and cognitive outcomes throughout the glycaemic range. Since HbA1c measures chronic hyperglycaemia more reliably than random/ fasting blood glucose testing alone, the availability of this measure in the Biobank cohort provides a new opportunity to investigate the impact of glycaemia on health outcomes. UK Biobank has large numbers of participants with baseline glycaemic status and incident measures of disease, thus providing a unique and powerful instrument to address these questions. By defining the role of sub-clinical hyperglycaemia in cardiovascular disease, cancer and cognitive decline, we aim to improve the prevention and diagnosis of these diseases, in line with UK Biobank?s core goals. To address these aims, we will require access to data only, to include the following health outcomes records: HES, GP data, cancer and death registers. Measures of HbA1c, socio-demographics, lifestyle factors, anthropometry, cardiovascular risk factors (e.g. blood pressure and cholesterol) and medical and drug history from baseline will all be required. We will use statistical models to show how baseline HbA1c relates to the health outcomes of interest, what other factors influence these associations and whether associations are different in population sub-groups. We intend to use the full dataset of approximately 500,000 people. Outcomes are often compared in populations with and without diabetes, but the validity of this binary classification and widely used cut-off points is not established. Including people with no diabetes as well those with treated and untreated diabetes offers an opportunity to disentangle effects of diabetes treatments on disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/chronic-high-impact-pain-and-uk-biobank-presentation-transitions-and-targets-for-intervention-chipp

Chronic High Impact Pain and UK Biobank: presentation, transitions, and targets for intervention (CHIPP)

Last updated:
ID:
98481
Start date:
19 July 2023
Project status:
Current
Principal investigator:
Professor Danielle van der Windt
Lead institution:
Keele University, Great Britain

BACKGROUND
Nearly half of UK adults have pain in their muscles or joints lasting longer than three months (chronic pain). While most people manage well, about 25% of people have pain that has far-reaching impacts on their lives, leading to disability, distress, social isolation, and high healthcare needs. It is not clear why some people experience such ‘high impact chronic pain’ whereas others don’t.

AIMS OF THE STUDY
1. Understand the different ways in which pain can impact on people’s lives, and agree better definitions of high impact chronic pain.
2. Investigate the reasons why chronic pain affects some people more than others, and why this can change over time. The study will look at:
a. psychological factors, such as mood, attention, memory, ability to problem-solve
b. trauma or impactful events that may occur at different times in people’s lives (such as a fracture, bereavement, or new illness)
3. Identify selfcare or treatment options that can reduce the influence of such factors in people with chronic pain.

RESEARCH PLAN
We will use data from UK Biobank to investigate chronic high impact pain. UK Biobank is a large-scale database, containing detailed genetic and health information from half a million UK participants. The data can be used by approved researchers and scientists from across the world, who carry out research in a wide range of health problems and diseases.
UK Biobank participants were aged 40-69 years when data collection started in 2006-2010. In 2019, 173,000 participants completed a questionnaire on the nature and impact of chronic pain and a second pain questionnaire will be sent out to in 2023. We will analyse data on pain, mental health, sleep, psychological factors, general health, healthcare use, and social factors to answer the research questions.

PATIENT AND PUBLIC INVOLVEMENT IN CHIPP
The aims of the research have been agreed together with public contributors, who shared their life stories, and suggested factors that may explain the impact of chronic pain. Involvement can be flexible depending on people’s interests, but may include
– helping to generate definitions for chronic pain with high impact
– prioritising factors to be included in the analysis
– reviewing self care and treatment options that may be suitable for people with chronic pain
– helping to interpret findings from the analysis
– formulating key messages and dissemination of findings


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/chronic-inflammatory-disorders-and-cardiovascular-disease

Chronic inflammatory disorders and cardiovascular disease

Last updated:
ID:
13933
Start date:
1 September 2015
Project status:
Closed
Principal investigator:
Dr Alexandru Dregan
Lead institution:
King's College London, Great Britain

Our recent findings published in Circulation indicated an association between several inflammatory disorders with cardiovascular disease (CVD). The study used primary care data with incomplete information on traditional vascular risk factors (ie hypertension, cholesterol) and demographic characteristics. In this study we aim to explore the possibility that the prevalence of inflammatory disorders and their association with incident CVD and mortality events vary across different deprivation and ethnic groups. Secondly, in a subsample of participants (N=190,000) we will test the hypothesis that inflammatory biomarkers and regulatory factors are associated with markers of atherosclerosis (arterial stiffness) and clinical outcomes. Our research is funded by the NIHR Biomedical Research Centre at Guy?s and St Thomas?. We aim to develop our expertise to utilising and analysing the UK Biobank data. The planned analyses will provide new understanding about the role of inflammation in CVD risk across different population subgroups. These findings will help identify the need for more targeted preventative measures. The use of inflammatory and CVD biomarkers will highlight potential mechanisms through which these inflammatory conditions may impact on CVD risk. The findings will facilitate enhanced risk prediction modelling and identify potential therapeutic targets for inflammatory disease patients. In the full UK Biobank cohort, baseline data on inflammatory disorders will be used to assess their prevalence and relationship with subsequent risk of CVD and mortality in subgroups defined by their deprivation and ethnicity. Baseline vascular risk factors (ie smoking, hypertension, cholesterol, diabetes, BMI) will be used as covariates. Additionally, the relationships between inflammatory biomarkers (ie CRP, rheumatoid factor) with inflammatory disorders, as well as with CVD and CVD-related biomarkers (ie arterial stiffness) will be estimated to explore mechanisms through which inflammation influences CVD risk. The impact of genetic regulators of inflammation biomarkers (IRF-5) will be explored. The analyses of inflammatory disorders and risk of CVD and mortality events will be conducted on the full cohort, except those with established CVD at baseline. When available in 2016, the relationships between measured inflammatory biomarkers with CVD will be conducted on the full cohort, except those with established CVD at baseline. The relationships between measured inflammatory biomarkers with CVD-biomarkers will be conducted on the subsample of patients (N=190,000) with a measure of arterial stiffness. We would like to request access to the genotype and primary care data when available, to identify incident cases of disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/chronic-ingestion-of-high-dose-proton-pump-inhibitors-ppis-and-development-of-pancreatic-cancer

Chronic Ingestion of high dose proton pump inhibitors (PPIs) and development of pancreatic cancer

Last updated:
ID:
63113
Start date:
16 June 2021
Project status:
Closed
Principal investigator:
Dr Jill P Smith
Lead institution:
Georgetown University, United States of America

The incidence of pancreatic cancer has more than doubled in the last 2 decades and in spite of our improved technology and therapeutics, the 5-year survival is still at a stagnant 7%. A protein called gastrin has been shown to stimulate growth of pancreatic cancer. The normal role of gastrin is to control acid production in the stomach. When people take certain medicines that decrease stomach acid for ‘acid reflux disease’, gastrin levels can increase. The medicines that are associated with increasing blood gastrin levels are called proton pump inhibitors (PPIs), such as Omeprazole. The reasons for the increased frequency of pancreatic cancer over the past 2 decades are unknown. We are hypothesizing that the increase may be related to the over use of these medications (PPIs) that increase gastrin. In this analysis we will examine the incidence of pancreatic cancer among those taking these medications and those not taking PPIs.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/chronic-pain-and-health-related-quality-of-life-a-population-based-study

Chronic pain and health-related quality of life: a population-based study

Last updated:
ID:
84853
Start date:
24 May 2022
Project status:
Current
Principal investigator:
Dr Nigel Armfield
Lead institution:
University of Queensland, Australia

Chronic pain, that is pain of greater than 3 months duration is a global public health problem. Previous studies have estimated that up to 43.5% of people experience some form of chronic pain, with 10% to 14% experiencing moderate to severe pain. Chronic pain is costly; annual costs have been estimated at up to 635 billion US dollars in the USA, and 200 billion Euros in Europe. The burden of chronic pain has been studied, typically through small-sized studies of individual conditions or pain locations, or by using data that have been aggregated and do not offer the detail that may be gained from a large study using individual participant data.

Our study will provide new and important knowledge about the burden of chronic pain at a population level. The new information will be useful to inform policy, decision making, and the evaluation of future interventions to improve the lives of those with chronic pain.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/chronic-pain-c-reactive-protein-vitamin-d-psychosocial-lifestyle-factors-and-medical-comorbidities-a-study-using-uk-biobank-data

Chronic pain, C-reactive protein, Vitamin D, psychosocial & lifestyle factors and medical comorbidities – a study using UK Biobank data

Last updated:
ID:
69067
Start date:
25 January 2021
Project status:
Closed
Principal investigator:
Dr Scott Francis Farrell
Lead institution:
University of Queensland, Australia

This study aims to determine if blood concentration of an inflammatory marker (C-reactive protein, CRP) and an anti-inflammatory hormone (Vitamin D) are associated with presence of long-term pain. It also aims to assess the influence of demographic, lifestyle, psychosocial factors and medical conditions on the relationships between long-term pain and CRP/Vitamin D.

Long-term (chronic) pain conditions are a major health challenge, with conditions like lower back pain and neck pain being the leading cause of disability in most countries worldwide. Our current approaches to treat chronic pain generally demonstrate modest effects. This may be due to our partial understanding of the biological and psychosocial factors underlying many chronic pain conditions.

Blood concentrations of CRP and Vitamin D appear to be associated with chronic pain. Clinical studies have found raised CRP or reduced Vitamin D in the blood of people with various chronic pain conditions. This could indicate that these blood markers are involved in the development or maintenance of chronic pain, which may have clinical significance for patient management. However, raised CRP and reduced Vitamin D are also associated with various demographic, psychosocial, lifestyle and medical factors. Given that chronic pain conditions are also commonly associated with similar demographic, psychosocial, lifestyle and medical factors, we therefore do not know if raised CRP and reduced Vitamin D are actually related to chronic pain conditions, or if CRP/Vitamin D levels in people with chronic pain simply reflect the influence of other psychosocial or medical factors.

Our proposed project using UK Biobank data will take approximately 18 months. We will examine blood concentrations of CRP and Vitamin D in people with chronic back, neck/shoulder, hip, knee, widespread and multisite pain, as compared with people with no pain. We will statistically adjust for demographic, psychosocial, lifestyle and medical factors, to determine whether raised CRP and reduced Vitamin D are independently associated with chronic pain, or alternatively, if blood CRP and Vitamin D concentration can simply be explained by demographic and psychosocial factors etc.

This study will improve our knowledge of the biological and psychosocial factors underpinning chronic pain conditions. This advanced understanding may guide patient assessment and could inform selection and development of new treatments for chronic pain.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/chronic-pain-in-shiftworkers-the-role-of-sleep-circadian-rhythms-and-the-autonomic-nervous-system

Chronic Pain in Shiftworkers: the Role of Sleep, Circadian Rhythms and the Autonomic Nervous System

Last updated:
ID:
40556
Start date:
28 September 2018
Project status:
Closed
Principal investigator:
Dr Kun Hu
Lead institution:
Brigham and Womens Hospital, United States of America

Chronic pain is pain that lasts longer, or is more severe than expected, deeply impacts not only those affected, but also family, friends and caregivers. Unfortunately, this is a common problem but effective treatments are few and far between. Understanding who is at risk so we can help minimise suffering becomes vital. We know that pain has both physical and psychological parts to it. The body has a natural ‘clock’ that cycles over roughly 24 hours, controlling vital functions that we are not even aware of (for example the autonomic nervous system). If this clock is disrupted, there is also both physical and psychological consequences. This project will look at the relationship between disruptions to the clock (as happens during shiftwork) and the development of chronic pain. We will also see if sleep disruption and the autonomic nervous system affects this relationship . We have also developed techniques that can examine heart rate tracings to reveal more information than just how fast the heart is going which we will test to see if they predict your likelihood to developing chronic pain. The public health impact is understanding who and how people develop such a debilitating disease – this may guide us to finding news ways to prevent or treat chronic pain.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/chronic-pain-prognostication

Chronic pain prognostication

Last updated:
ID:
83292
Start date:
29 April 2022
Project status:
Closed
Principal investigator:
Dr Scott Thompson
Lead institution:
University of Minnesota Twin Cities, United States of America

A curious thing about chronic pain is that two people can suffer the same acute trauma or have the same age-related structural deterioration but one person will develop chronic pain and the other will not. Frustratingly, the underlying reasons are unclear. This, in part, stems from the complex nature of chronic pain which is influenced by biological, psychological, and social factors that prime the body to intensify and maintain pain. These factors include things like smoking, alcohol consumption, obesity, exercise habits, sleep, nutrition, employment status, age, sex, cultural background, history of trauma and heritable factors. While these factors have been studied by many researchers, the diversity and complexity of the interaction between these factors have made it difficult to reliably identify sets of factors that predict chronic pain outcomes. To create a tool that is clinically relevant, all of the aforementioned factors need to be considered simultaneously on a group large enough to support modern analysis methods. The UK Biobank is an unprecedented health trajectory study which enable this approach with baseline (2006-2010) information about the participants followed by a comprehensive pain survey in 2019. The application of modern analytical techniques allows for all of the factors mentioned above to be considered at the same time to identify people with high risk for i) developing chronic pain, ii) developing high-impact pain, and iii) recovering from pain (our study aims). Given the complex nature of these analysis, we anticipate the project duration to be 3 years. These results are expected to have a positive public health impact because they are likely to provide a strong evidence-based framework for future clinical guidelines for reducing chronic pain development, severity, and duration.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/chronic-venous-disease-an-underestimated-risk-factor-for-cardiovascular-health

CHRONIC VENOUS DISEASE: AN UNDERESTIMATED RISK FACTOR FOR CARDIOVASCULAR HEALTH?

Last updated:
ID:
358626
Start date:
11 March 2025
Project status:
Current
Principal investigator:
Professor Emilton Lima Junior
Lead institution:
Universidade Federal do Paraná, Brazil

The objective of this research is to understand the relationship between venous disease and cardiovascular events such as stroke, heart attack, and their associated risk factors, including high blood pressure, diabetes, and smoking habits. Increasing evidence indicates a significant connection between these conditions. Substances secreted by cells in the arterial system are similar, if not identical, to those produced by cells in the venous walls. This suggests a potential crosstalk of risk factors throughout the circulatory system, where a chronic inflammatory state in the veins could play a crucial role in severe cardiovascular diseases, including those affecting cardioimmunometabolic health.
At rest, blood takes approximately one minute to travel through the body’s network of arteries and veins, highlighting the ongoing communication between the heart, arteries, and veins in both healthy and diseased states.
The study is expected to last for 36 months. Investigating this relationship is important because venous disease might be a preventable risk factor for cardiovascular events. Understanding this connection could lead to improved prevention and treatment strategies, ultimately reducing the incidence of heart attacks, strokes, and related health issues.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/chronotype-and-resting-state-connectivity

Chronotype and resting state connectivity

Last updated:
ID:
71741
Start date:
16 June 2021
Project status:
Closed
Principal investigator:
Dr Ray Norbury
Lead institution:
Brunel University London, Great Britain

Humans display near 24 hour circadian rhythms that drive our preferences for wakefulness, periods of activity and sleep. Some people naturally prefer to rise early and go to bed early (morning larks) whereas others prefer to rise late and go to bed late (night owls) and increasing evidence suggests that these preferences in sleep and wake timing may impact aspects of cognitive function and physical and mental health. The human brain has a number of well recognised intrinsic functional neuronal networks that support a diverse range of functions including different types of memory and how we process emotional information and these can be explored using a particular type of brain scan called Resting Functional Magnetic Resonance Imaging or Resting FMRI. In this project, we will investigate Resting FMRI in morning larks and night owls using UK Biobank brain imaging data combined with numerous measures of lifestyle. We expect to demonstrate significant differences in resting functional connectivity between morning larks and night owls which may underlie the differences in emotional processing and some aspects of memory observed in these groups. We believe this is important for understanding how circadian rhythms impact on thinking and feeling and could inform interventions, such as flexible start times for work or university that better match individual circadian rhythm and may benefit individuals with a more evening orientation.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/chronotypes-impact-on-diurnal-patterns-of-sedentary-and-physical-activity-and-mortality-in-non-exercisers-a-prospective-study

Chronotype’s Impact on Diurnal Patterns of Sedentary and Physical Activity and Mortality in Non-Exercisers: A Prospective Study

Last updated:
ID:
161289
Start date:
26 March 2024
Project status:
Current
Principal investigator:
Dr Jong-Hwan Park
Lead institution:
Pusan National University Hospital, Korea (South)

Aims
The chief aim of this investigation is to clarify how chronotype impacts daily cycles of physical exertion and inactivity, specifically in individuals who don’t engage in routine exercise. An additional aim is to investigate the potential connections between these daily patterns and ethical considerations, here referred to as “morality”. The focus of the study encompasses the impact of chronotypes on sedentary behaviors and activity routines, and their correlation with various health issues such as heart disease, cancer, and mental health in non-exercisers.

Scientific Justification
From a scientific standpoint, chronotype is understood as an individual’s natural preference for the timing of their activities and has been associated with significant health outcomes. Prior research, particularly animal studies, has consistently demonstrated the metabolic effects of activity timing. However, human studies have produced varied results. This study intends to fill this gap in knowledge by concentrating on non-exercisers, a group particularly vulnerable to health problems like heart diseases, cancers, mental disorders, and metabolic diseases.

Project Timeline
The investigation is planned for a span of three years, during which time-series data will be gathered from subjects. This dataset will include variables like daily activity metrics, sleep cycles, and general health indicators. Sophisticated statistical techniques, such as k-means cluster analysis and latent profile identification, will be utilized for data scrutiny.

Implications for Public Health
The prospective public health ramifications of this investigation are noteworthy. By clarifying the nexus between chronotype, daily activity cycles, and health metrics, the study aims to guide specialized public health initiatives. Specifically, the insights could aid in crafting health campaigns aligned with chronotype, thereby boosting their effectiveness. Additionally, the study could enrich the emerging domain of personalized healthcare by empowering medical professionals to offer lifestyle guidance based on chronotype.

Summary
In essence, this investigation seeks to offer an exhaustive understanding of how chronotype shapes daily cycles of exertion and inactivity among non-exercisers, and to probe its ethical implications. The study has considerable potential for both academic exploration and shaping public health strategies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/circadian-rhythm-disruption-lifestyle-and-liver-diseases

Circadian rhythm disruption, lifestyle, and liver diseases

Last updated:
ID:
287670
Start date:
18 December 2024
Project status:
Current
Principal investigator:
Dr Yikyung Park
Lead institution:
Washington University in St. Louis, United States of America

Liver disease, such as fatty liver and liver cancer, is a rapidly growing illness worldwide. Obesity and metabolic problems are known to increase the risk of these liver diseases. However, we have limited knowledge about the role of other lifestyle factors, specifically those that disrupt our internal body clocks. When our internal body clocks are thrown off, mostly due to changes in our daily routines and environments, it can lead to metabolic issues that contribute to fatty liver diseases and potentially liver cancer as well. Therefore, this study aims to investigate whether there exist image-based marker and/or lifestyle factors that disrupt our body clocks are linked to a higher risk of liver diseases. This project will take approximately 36 months to complete.

Considering the escalating burden of liver diseases, it is crucial to focus on prevention efforts. This study will identify new modifiable risk factors that can be targeted through lifestyle interventions, making a significant contribution to disease prevention.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/circadian-rhythm-psychosocial-well-being-and-cognitive-health

Circadian rhythm, psychosocial well-being, and cognitive health

Last updated:
ID:
218723
Start date:
15 January 2025
Project status:
Current
Principal investigator:
Dr Xianghe Zhu
Lead institution:
Wenzhou Medical University, China

Circadian rhythm disturbances, which include sleep apnea, sleep difficulty, altered sleep pattern and duration, among others, are not only prevalent public health issues per se, but are also associated with changes in cognitive function and risk of dementia. At the meantime, changes in both circadian rhythm and cognitive health are imbedded in one’s psychosocial environment. However, the associations between circadian rhythm and cognitive health, particularly the mechanisms and the role of psychosocial factors, are not fully understood.

We propose to examine the associations between circadian rhythms with various dimensions of cognitive health, including cognitive performance, age-related cognitive decline, risk of Alzheimer’s Disease and related Dementias, among others. We are particularly interested in the roles of psychological and social well-being as well as biomarkers in these associations.

The project will take approximately 3 years.

For public health, findings of the proposed study will help to elucidate the potential influences of circadian rhythms and sleep disturbances on different aspects of cognitive health as well as the mechanisms. In particular, for healthy aging, the study may provide evidence for early prevention and intervention for cognitive decline and impairment through lifestyle and psychosocial approach.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/circulating-glp-1-related-biomarkers-in-heart-failure-prognostic-value-of-proglucagon-and-dpp4

Circulating GLP-1-Related Biomarkers in Heart Failure: Prognostic Value of Proglucagon and DPP4

Last updated:
ID:
724929
Start date:
7 April 2025
Project status:
Current
Principal investigator:
Dr Arzu Kalayci
Lead institution:
Baim Institute for Clinical Research, United States of America

Heart failure (HF) is a complex syndrome characterized by impaired ventricular function and heterogeneous phenotypes, including HF with preserved ejection fraction (HFpEF), HF with mildly reduced ejection fraction (HFmrEF), and HF with reduced ejection fraction (HFrEF). HFpEF is driven by diastolic dysfunction and systemic inflammation, while HFrEF is characterized by systolic dysfunction and adverse remodeling, whereas HFmrEF represents an intermediate phenotype. These subtypes have distinct pathophysiologies, risk factors, and treatment responses, highlighting the need for targeted therapeutic strategies. Glucagon-like peptide-1 receptor agonists (GLP-1 RAs) and dipeptidyl peptidase-4 (DPP4) inhibitors have demonstrated cardiometabolic benefits, yet their therapeutic role in HF remains uncertain. Importantly, the impact of circulating GLP-1-related biomarkers on HF outcomes and their potential role in predicting therapeutic response remain unknown.
This study aims to analyze circulating levels of proglucagon (GLP-1 precursor) and DPP4 (GLP-1 degrading enzyme) levels in UK Biobank participants with HFpEF, HFmrEF, and HFrEF to assess their associations with adverse cardiovascular outcomes, including HF hospitalization, cardiovascular mortality, and all-cause mortality. Through stratification of patients by HF subtype, we aim to determine whether these biomarkers provide prognostic value and help identify patients most likely to benefit from incretin-based therapies within specific HF subgroups. Findings from this study could provide foundational evidence, bridging a critical knowledge gap and offering novel mechanistic insights into the GLP-1/DPP4 pathway in HF subtypes, potentially informing patient selection for future therapeutic interventions. These biomarker-driven insights may advance precision medicine approaches, help shape future clinical trials of incretin-based therapies, and guide the use of GLP-1 RAs or DPP4 inhibitors in HF management.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/circulating-plasma-metabolites-lifestyle-adiposity-and-biochemical-exposures-and-risk-of-major-chronic-diseases-and-their-co-occurence

Circulating plasma metabolites, lifestyle, adiposity, and biochemical exposures and risk of major chronic diseases and their co-occurence

Last updated:
ID:
79696
Start date:
4 May 2022
Project status:
Current
Principal investigator:
Dr Christopher Papandreou
Lead institution:
Institut d'Investigació Sanitària Pere Virgili (IISPV), Spain

The prevalence of cardiovascular diseases (CVD), type 2 diabetes (T2D), cancer and dementia/Alzheimer’s disease (AD) has risen partially due to the aging of the Western populations and also to increases in shared risk factors. Although increasing evidence has linked unhealthy lifestyle, adiposity and abnormal biochemical measures to the risk of these diseases, the underlying mechanisms remain unclear yet. Metabolomics, through a systematic evaluation of small-molecule metabolites in biological samples such as blood, may help to study these exposures and health outcomes because it integrates information from genetic and environmental factors. To our knowledge, few studies have identified early biomarkers of common diseases and even less evidence exist on metabolic signatures of a number of exposures, simultaneously, in relation to diseases. Metabolomics, would allow the discovery of biomarkers and identification of novel metabolic pathways of exposures involved in the development of common diseases and thus advance our understanding on the underlying biological mechanisms by which the exposure is acting. We propose, for the duration of three years, to perform a prospective analysis by using large-scale metabolite, genetic, exposure, and clinical data from the UK Biobank. In this project, we will: 1) examine associations between baseline metabolites and risk of CVD, T2D, several types of cancer and dementia/AD; 2) investigate associations of lifestyle, circadian rhythm, adiposity and biochemical exposures with risk of these diseases; 3) try to identify metabolite signatures of each exposure and associate them with the risk of these diseases; 4) estimate the extent to which the identified metabolite signatures mediate the associations between the exposures and diseases; 5) examine causality of the associations between the identified metabolites or metabolite signatures and risk of diseases; 6) investigate the role of the identified exposures, metabolites and metabolite signatures in the risk of progressing from one of these diseases to multimorbidity. The results of this project will be useful to elucidate the underlying mechanisms in the relationship between exposures to highly prevalent diseases risk. The identified metabolites and metabolomics signatures related with these diseases and their co-occurrence could be used as target for prevention or treatment of both single disease and multimorbidity.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/clarification-of-causes-of-multifactorial-diseases-by-phenotypic-clustering-using-uk-biobank-data

Clarification of causes of multifactorial diseases by phenotypic clustering using UK Biobank data

Last updated:
ID:
74297
Start date:
18 March 2022
Project status:
Current
Principal investigator:
Professor Shinichi Kuriyama
Lead institution:
Tohoku University, Japan

The causes of allergic diseases, cardiovascular disease risk factors, depression, and cancer are not sufficiently clear in terms of both genetic and environmental factors. One of the reasons for this may be that these diseases are syndromes composed of multiple diseases rather than a single disease. The purpose of this study is to divide these syndrome-like diseases into more similar subgroups by analyzing the phenotypes of these diseases in terms of symptoms and severity.

We have already succeeded in dividing patients with autism spectrum disorders with potentially similar genetic factors by applying artificial intelligence analysis technology. Based on the progress of this study, we aim to divide allergic diseases, cardiovascular disease risk factors, depression (including major depressive disorder, bipolar disorder, and anxiety disorder), and cancer into subgroups to make these syndromes into more similar disease concepts during a rolling 3-year period of our project. These findings will help to elucidate the causes of diseases from which many people suffer and for which many causes have been postulated.

On the basis of the present study, individual risks can be estimated using the genetic factors identified in the subgroups, while taking into account environmental factors. Knowing these risks will help to devise optimal treatment interventions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/classification-and-automatic-detection-of-oct-layer-segmentation-failure-modes

Classification and automatic detection of OCT layer segmentation failure modes

Last updated:
ID:
41041
Start date:
25 June 2018
Project status:
Closed
Principal investigator:
Dr Ryan Amelon
Lead institution:
IDx, LLC, United States of America

Automated AI diagnostic solutions are expected to properly function and yield accurate results all of the time, but no solution will ever have perfect accuracy. It is important to consider failure modes of automated diagnostics components, both when they fail and how. The detection and proper characterization of these failure modes can make automated diagnostics safer and more efficient.
Glaucoma in the retina is characterized, in part, by a loss of retinal nerve fiber layer thickness. Optical coherence tomography (OCT) imaging results in a three-dimensional view of the retina which allows for measurement of retinal nerve fiber layer thickness. The usual first step in detecting Glaucoma using OCT is segmentation of the layers of the retina. Typical assessment of these algorithms consider the accuracy of the segmentation, which is important when considering algorithm utility. However, most assessments ignore, at a patient level, how often the layer segmentations fail and what is causing the failure. For automated algorithms to become common-place, it must be able to both perform the task it was trained for as well as determine when the task cannot be adequately performed. Further, if an algorithm can determine why it failed, a patient visit may be correctly salvaged by specifying the necessary follow-up steps required for an accurate result.
This work will shed light on the mechanisms of layer segmentation failures that developers should be aware of, how common such failures may occur, and the ability to which they may be automatically detected. This study will provide a proof-of-concept failure mode assessment that will increase device safety and efficacy by correctly identifying algorithm failures and suggest follow-up measures.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/classification-and-prediction-of-cardiovascular-disease-using-traditional-and-psychological-risk-factors

Classification and Prediction of Cardiovascular Disease Using Traditional and Psychological Risk Factors

Last updated:
ID:
799839
Start date:
10 June 2025
Project status:
Current
Principal investigator:
Mr Tushar Saha
Lead institution:
Central Institute of Technology (CIT), Kokrajhar, India

Cardiovascular disease (CVD) is one of the leading causes of death worldwide. While risk factors like age, body mass index (BMI), blood pressure, cholesterol, and smoking are well-known, recent studies suggest that mental health issues such as depression, anxiety, stress, and mood swings may also play a role in developing CVD.

This project aims to study the combined effect of physical and psychological factors on heart disease using data from the UK Biobank. We want to find out if adding mental health data can improve how well we predict a person’s risk of getting CVD.

Our main goals are:
a) To understand how mental health conditions relate to heart disease
b) To build machine learning models that use both physical and mental health information to predict CVD risk

This research could help create more complete and accurate health risk assessments, leading to better prevention and care strategies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/classification-and-prediction-of-disease

Classification and Prediction of Disease

Last updated:
ID:
42225
Start date:
21 February 2019
Project status:
Closed
Principal investigator:
Professor Lawrence Sirovich
Lead institution:
The Rockefeller University, United States of America

We have made a careful study of the informational content of an individual’s DNA, and the potential mutations which may appear. We have been able to link mutations to disease for seven diseases (Bipolar Disorder, Coronary Artery Disease, Crohn’s Disease, Hypertension, Rheumatoid Arthritis, Type-1 Diabetes, and Type-2 Diabetes). These deliberations have been put into the form of classifiers of disease, and classifiers of wellness with respect to these diseases. The UK BioBank database offers us an opportunity to evaluate the new methods we developed on independent data. Since it is a low probability that any of the sequences in the WTCCC1 database are also present in the Bio Bank database, this offers a relatively blind test of the methods we are proposing. It is our contention that the rationale for this scientific effort is compelling. It is also our belief that we can make sufficient progress in the first year of this investigation to sensibly evaluate the novel approach. It is our belief that the public health aspect of our endeavors can be immense.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/classification-of-asthma-patients-and-identification-of-group-specific-genetic-variants

Classification of asthma patients and identification of group-specific genetic variants

Last updated:
ID:
56987
Start date:
14 January 2020
Project status:
Closed
Principal investigator:
Dr Han-Kyul Kim
Lead institution:
Kyung Hee University, Korea (South)

Asthma is a common respiratory disease with a prevalence of more than 5 percent of adults worldwide, requiring huge public healthcare cost burdens. Recently it has been known that asthma is a kind of clinical syndrome that shows heterogeneity of clinical phenotypes. This study aims to identify asthma group-specific genetic markers by the classification analysis of asthma patients and follow-up genome-wide association studies. Though most asthma can be treated by conventional bronchodilators, some need particular treatment such as long-acting !2 agonists or inhaled corticosteroids and even the expensive antibody-based drugs such as Anti-IgE or Anti-IL5 are required in severe asthma for improvement in symptoms. To make it worse, effective drug control is often complicated by asthma heterogeneity in clinical phenotypes. Indeed, asthma is a heterogeneous disease with individually distinct clinical features. Therefore, asthma classification is important before the selection of the right clinical care. Previously classification of asthma patients was successfully carried out according to clinical phenotypes. However, this classification does not provide molecular information about the heterogeneity, and also underlying biology of each group. In order to understand the molecular mechanism, we will classify over 60,000 asthma patients of UK Biobank into groups using the clinical variables related to asthma and then identify group-specific genetic markers. We will use diverse clinical measurements such as lung function, onset age, blood cell counts, smoking, and atopy status for the classification. Genetic markers unique to asthma group may help classify patients in addition to the classification of asthma patients by clinical phenotypes. Moreover, these genetic markers can be useful for delineating molecular pathways of asthma group that will help severe asthma patients select the right drug based on biological markers such as Anti-IgE and Anti-IL5. Ultimately, this study will provide tools for the modeling of prediction of asthma grouping as well as the precision medicine through tailored health care to alleviate patients’ economic and physical burdens. This study may take three years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/classification-of-major-depressive-disorder-from-brain-fmri-scans-using-machine-learning-techniques

Classification of Major Depressive Disorder from Brain fMRI Scans using Machine Learning Techniques

Last updated:
ID:
203635
Start date:
30 April 2024
Project status:
Current
Principal investigator:
Ms Reet Khare
Lead institution:
Pune Institute of Computer Technology, India

Our research project aims to develop a better understanding of Major Depressive Disorder (MDD) and improve early detection and diagnosis through advanced machine learning techniques. We want to figure out which machine learning method works best among Support Vector Machine, LightGBM, and Convolutional Neural Networks (CNN), and CNN with Transfer Learning.

The scientific rationale behind our project is straightforward: by harnessing the power of machine learning, we can analyze large amounts of data to identify subtle patterns and indicators of MDD. This could lead to earlier detection and intervention, ultimately improving outcomes for individuals struggling with this condition.

The project duration is estimated to span 6-8 months, depending on the complexity of the analysis and the availability of data.

The potential public health impact of our research is significant. By improving early detection and diagnosis of MDD, we can help healthcare professionals intervene sooner, potentially preventing the worsening of symptoms and reducing the overall burden of the disorder on individuals and society. Additionally, our findings could inform the development of more effective screening tools and treatment strategies for MDD, leading to better outcomes and quality of life for those affected.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/classification-of-mental-disorders-using-multi-modal-data

Classification of mental disorders using multi-modal data

Last updated:
ID:
74831
Start date:
5 July 2023
Project status:
Current
Principal investigator:
Dr Bo Cao
Lead institution:
University of Alberta, Canada

It has been demonstrated that the biological data (e.g., genetic and neuroimaging data) from the UK Biobank can provide valuable information in diagnosis prediction of mental disorders. However, it is less clear whether non-biological data (e.g., behavioral, cognitive and clinical assessments and health records) is as predictive for mental disorders. If so, will the biological data further enhance the accuracy of mental disorder prediction when they are combined with non-biological data? In this study, we aim to address the two questions by combining the various forms of data from the UK Biobank and applying advanced machine learning algorithms to this combined data. This project is expected to take 5 years to complete. Through this project, we will develop a computational tool for individualized mental disorder identification, which may improve the diagnosis and facilitate better understanding of mental disorders.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/classification-of-obesity-based-on-different-disease-susceptibility

Classification of obesity based on different disease susceptibility

Last updated:
ID:
454980
Start date:
27 March 2025
Project status:
Current
Principal investigator:
Mr Dewen Dong
Lead institution:
China Medical University P.R.C, China

Background:John Speakman’s article “Open Questions about the Causes of Obesity” published in the journal Science in 2023 suggests that obesity should be classified more rationally. Based on this, we also believe that the current classification of obesity is not detailed enough, and it is known that obesity often has complications such as cardiovascular disease and cerebrovascular disease. Based on this background, we believe that it is possible to combine obesity with diseases, so as to classify them according to comorbidities.
Principle:The common proteins between obesity and other diseases were searched, and they were classified by similar protein expression.
Method!COX survival analysis was performed based on UKB population data and UKB-PPP data, including age, gender, height, smoking, diet and other covariates, and proteins related to obesity and other diseases were screened. Finally, enrichment analysis was performed on other disease protein results and obesity protein results, and common proteins were screened as markers of susceptibility to different diseases.
Data:I need UKB whole queue data and UKB-PPP whole data.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/classifying-prodromal-stage-of-lewy-body-disease-by-symptoms-and-underlying-factors

Classifying prodromal stage of Lewy body disease by symptoms and underlying factors.

Last updated:
ID:
293613
Start date:
23 October 2024
Project status:
Current
Principal investigator:
Dr Daigo Tamakoshi
Lead institution:
Nagoya University, Japan

Lewy body disease (LBD) , including Parkinson’s disease (PD) and dementia with Lewy bodies(DLB) , is characterized by Lewy bodies within central and peripheral neuronal cells. Even before the disease onset, LBD patients often present non-motor symptoms such as autonomic disorder, REM sleep behavior disorder, olfactory dysfunction, and depression as prodromal symptoms, reflecting the propagation of Lewy bodies and neurodegeneration. Elucidating the pathogenesis of the prodromal phase is useful for early diagnosis and development of disease-modifying therapies.

Most of today’s research on the prodromal phase of LBD has been conducted in patients with idiopathic REM sleep behavior disorder (iRBD) because most of the patients with iRBD will develop neurodegenerative disease such as LBD in the future. However, only 20-30% of PD patients present RBD, implying diversity of LBD. The prodromal symptoms and disease progression vary from individual to individual and it supposed to be caused by different pathophysiological background.

The aim of this study is to clarify the diversity of the prodromal pathophysiology of LBD, and constructing a model to predict disease progression and to develop novel disease-modifying therapies. We intend to classify LBD by analyzing the relationship between clinical information such as the various prodromal manifestations and the disease progression after disease onset, underlying genetic, biological, environmental, and lifestyle factors, and omics data.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/classifying-retinopathies-from-fundus-images-and-optical-coherence-tomography-data-using-deep-learning-methods

Classifying retinopathies from fundus images and optical coherence tomography data using deep-learning methods

Last updated:
ID:
95789
Start date:
16 February 2023
Project status:
Current
Principal investigator:
Mr Che-Ning Yang
Lead institution:
National Taiwan University, Taiwan, Province of China

This study trying to establish a deep learning system for retinopathies recognition, including polyarteritis nodosa retinopathy and diabetic retinopathy.
Diabetic retinopathy was caused by damage to the blood vessels of the retina due to consistently high blood sugar. It might cause no symptoms or only mild vision problems initially, but it can lead to blindness in end stages. Therefore, early diagnosis and timely treatment were necessary. Besides, polyarteritis nodosa (PAN) is a medium vessel vasculitis that may affect the eyes and the central neural system. About 10% of patients suffered retinopathy due to the disease progression, which may damage their vision severely. What’s more, PAN retinopathy and diabetic retinopathy were caused by different etiology, making them present different morphologies in the fundus.
Because of the above reasons, this study is going to develop deep-learning systems, which use algorithms and statistical models to analyze pictures and data, for detecting retinopathies, and the project duration would be expected as 36 months in total. The research not only investigates the variety of morphologies in the fundus in different retinal diseases but also a great aim for early detection of retinopathy and timely treatment. In addition, due to its characteristics of being cheap, rapid, and easily obtained, the systems could become a nationwide routine screening for early detection and timely treatment, which improved patients’ outcomes with better preservation of vision.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/clinical-and-biological-relevance-of-somatic-mutations-in-genes-associated-with-autoinflammatory-disorders

Clinical and biological relevance of somatic mutations in genes associated with autoinflammatory disorders

Last updated:
ID:
82060
Start date:
9 August 2022
Project status:
Current
Principal investigator:
Dr Sinisa Savic
Lead institution:
University of Leeds, Great Britain

The main purpose of the immune system is to protect our bodies from infection with viruses, bacteria and parasites. There is a group of conditions known as autoinflammatory disorders (AID) in which part of the immune system acts in an uncontrolled fashion and attacks healthy body tissues. These conditions are not always easy to recognise since many characteristic symptoms of AID, such as intermittent fevers, are also seen in people who are suffering from an infection. This means that it can be very difficult to make a diagnosis. A genetic cause has been found for several AIDs and this has been hugely helpful in making the diagnosis and selecting the best treatment. However, there are many patients in whom an obvious genetic cause is unknown. This is particularly the case for people who first develop symptoms in adulthood (> 50%). We now know that in some of these patients the genetic changes were not present at birth, but developed at a point in later life. Such genetic changes are acquired in a similar way to the genetic changes which lead to cancer, but instead of causing cancer they cause an inflammatory condition. Based on preliminary studies, we believe that acquired AIDs are more common then previously thought. However, their exact prevalence is not known. We also do not know how long it takes to develop an AID from the point when the acquired genetic change is first detected. We plan to use genetic information from the UK biobank to determine how commonly these genetic changes are seen in otherwise healthy people, and then follow up a selected group for up to 5 years to determine if the genetic changes progress and eventually cause disease. It is important to be able to identify AID early because we now have excellent treatments available. With our work, we also hope to gain important insights into the chronic inflammatory diseases which are common as people age, because there is likely to be overlap between the acquired genetic changes seen in these conditions and AIDs.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/clinical-and-genetic-association-studies-of-the-development-treatment-and-prognosis-of-heart-failure-related-cardiovascular-diseases

Clinical and genetic association studies of the development, treatment, and prognosis of heart failure -related cardiovascular diseases

Last updated:
ID:
97332
Start date:
6 January 2023
Project status:
Current
Principal investigator:
Dr Julio D Duarte
Lead institution:
University of Florida, United States of America

Aims and rationale: Despite the advancement in HF treatment, HF remains a growingly common and incurable heart disease, with 5-year death rate standing about 50%. HF can develop and deteriorate from multi-layer reasons, but not every reason has been understood to a level where drug treatment can be developed to target that reason. The goal of our project is to better understand how HF develops and deteriorates from the perspective of its relationship with metabolic diseases, such as high blood pressure, high glucose, high cholesterol, and obesity. Understanding the details of disease development will help people to find effective treatments.
Project duration: We plan to complete the project within 3 years. Depending on the promises of the study results, we may extend the scope and /or duration of the project.
Public health impact: The treatment options for many types of HF and its resulted lung disease are limited to date. Our study will help to find key factors and genes involved in HF development and deterioration. These factors and genes may have the potential to become the targets for drug treatment. Our work will also help people to understand how some drugs that are used for metabolic diseases might also protect the heart. The knowledge would promote the search for HF treatments from available drugs.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/clinical-and-genetic-characterization-of-hypermobile-ehlers-danlos-syndrome-and-irritable-bowel-syndrome-and-in-the-uk-biobank-cohort

Clinical and genetic characterization of hypermobile Ehlers-Danlos Syndrome and Irritable bowel syndrome and in the UK Biobank cohort.

Last updated:
ID:
47591
Start date:
6 March 2020
Project status:
Closed
Principal investigator:
Qasim Aziz
Lead institution:
Queen Mary University of London, Great Britain

Aims
To find genetic variants associated with hypermobile EDS and IBS as well as hEDS-IBS overlap phenotypes through genome wide association study (GWAS) of well-phenotyped individuals in UK Biobank, UK’s largest cohort for genetic studies.

Scientific Rationale
Evidence suggests a link between connective tissue disorders like Ehlers-Danlos Syndrome (EDS) and gastrointestinal symptoms. A significant proportion of patients suffering with gastrointestinal (GI) symptoms also meet the criteria for hypermobile subtype of Ehlers-Danlos Syndrome (hEDS). GI symptoms in hEDS include both structural malformations like hiatus hernia, prolapse and functional GI symptoms like dysmotility, IBS, constipation, nausea, heartburn and others. In a recent study our group found that one-third of patients attending secondary care gastroenterology clinics meet the criteria for hEDS. Among these, prevalence of hEDS was approximately 40% in irritable bowel syndrome (IBS) patients and 50% in patients with functional dyspepsia. A similar study in USA, showed that 30.3% of their hEDS patients met the criteria for IBS. Although these and many other small studies suggest of a link between hEDS and IBS, there is a need to systematically study the genetic landscape as well as the clinical symptoms experienced by these patients in a large population cohort, in order to improve our understanding of pathophysiology in these two discrete conditions with substantial overlap in clinical symptoms.

Project duration
The tentative duration of project is 36 months.

Public health impact
The outcome of this research will help in establishing a genetic testing service to discriminate the hEDS-IBS patients from IBS alone. Providing a definite diagnosis to a category of patients hitherto either often categorized as unexplained medical disorders will be a major step in legitimizing their complaints, leading to earlier diagnosis and better management strategies leading to improved patient outcomes and quality of life and health care savings for the healthcare sector.

The outcome of the research will be shared with the scientific community through paper publication, conference presentation and engaging the international EDS society and its local chapter (EDS UK), International Foundation for Gastrointestinal Disorders and other organizations promoting research and awareness in IBS. Furthermore, medical magazines, and social media will also be used to disseminate the scientific data to a wider audience, including clinical and social practitioners.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/clinical-and-genetic-determinants-of-variation-in-glycaemic-response-and-trajectory-in-type-2-diabetes

Clinical and genetic determinants of variation in glycaemic response and trajectory in type 2 diabetes

Last updated:
ID:
20405
Start date:
2 December 2016
Project status:
Current
Principal investigator:
Professor Ewan Pearson
Lead institution:
University of Dundee, Great Britain

Patients with type 2 diabetes have variable response to treatment, and variable rates of progression of underlying disease. Response and progression are difficult to disentangle as progression encompasses underlying drug response. The research questions are:
What are the clinical, drug drug interaction and genetic predictors of drug response and how do these predictors differ between different diabetes drug treatments.
What is the variability in rate of diabetes progression between individuals, and what are the clinical and genetic predictors of variability in diabetes progression. How do non-diabetes drugs impact on diabetes progression? This project aims to improve the treatment of type 2 diabetes by gaining insight into the clinical and biological mechanisms that determine response to drugs and underlying diabetes progression. This will enable a stratified approach to therapy and the potential for development of novel therapies in this area. We will use the clinical data obtained from GP records in UK biobank to define how individuals with Type 2 diabetes respond to drug treatment, and how their diabetes progresses over time. We have developed statistical and genetic models for these outcomes within the Scottish health record data and will test and extend these in the large UK biobank dataset. Full cohort


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/clinical-and-molecular-features-of-metabolic-dysfunction-associated-steatotic-liver-disease-masld-and-associated-comorbidities

Clinical and molecular features of metabolic dysfunction-associated steatotic liver disease (MASLD) and associated comorbidities

Last updated:
ID:
677735
Start date:
21 April 2025
Project status:
Current
Principal investigator:
Dr Kara Wegermann
Lead institution:
Duke University, United States of America

Background and Aim: Metabolic dysfunction-associated steatotic liver disease (MASLD) affects approximately one-quarter of the world’s population. MASLD results in substantial morbidity, mortality, and healthcare expenditure. Our aim is to discover metabolomic and proteomic predictors of MASLD, define clinical and molecular features of MASLD in special populations, and describe metabolomic and proteomic predictors of comorbidities including cardiovascular disease in individuals with MASLD.

We plan to phenotype MASLD using hepatic steatosis index and fatty liver index, requiring waist circumference and basic laboratory values.

By utilizing comprehensive UK Biobank data, we aim to address the following research questions:
1) Metabolomics:
a. Can specific blood metabolite profiles predict presence or development of cardiovascular diseases in individuals with MASLD?
b. Can specific blood metabolite profiles predict MASLD in high-risk populations including people with HIV?
2) Proteomics:
a. Can specific plasma proteomic signatures predict the presence of hepatic steatosis?
b. Which proteomic markers can robustly predict liver-related events independently or in conjunction with other clinical data?
c. Can plasma proteomic signatures predict cardiovascular disease in individuals with MASLD?
3) Clinical Data:
a. How effectively can integrated models that combine clinical and proteomic data predict the presence of hepatic steatosis compared to traditional risk factors alone?


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/clinical-and-molecular-heterogeneity-of-kennedys-disease

Clinical and molecular heterogeneity of Kennedy’s disease

Last updated:
ID:
92363
Start date:
19 July 2023
Project status:
Current
Principal investigator:
Dr Matteo Zanovello
Lead institution:
University College London, Great Britain

Kennedy’s disease is an incurable, male-specific disease that affects the muscles and fat metabolism. The genetic cause of Kennedy’s disease is an expansion of a repeated DNA sequence in the Androgen Receptor gene. We recently discovered that the frequency of this DNA defect in the general population is 10 times more frequent than the reported Kennedy’s disease prevalence.
We wish to use the largest cohort to date, the UK Biobank, to explain the gap between the frequency of the genetic defect and disease prevalence.
This will be important for understanding the different symptoms of Kennedy’s disease in patients and the biology behind these. The findings from this work can be relevant not only for Kennedy’s disease but also for other disorders.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/clinical-and-prognostic-validation-of-a-new-diagnostic-criteria-of-sarcopenic-obesity-based-on-body-composition

Clinical and prognostic validation of a new diagnostic criteria of sarcopenic obesity based on body composition.

Last updated:
ID:
97619
Start date:
21 June 2023
Project status:
Current
Principal investigator:
Dr Vittoria Zambon Azevedo
Lead institution:
Sorbonne Université, France

The aims of this project are to establish an approach to diagnose a disease called sarcopenic obesity (SO). This is a condition whereby people with obesity (i.e. with increased adiposity) have a reduced muscle mass and reduced muscle function, a combination of factors which has many negative health consequences. This condition can worsen the course of many other diseases of the heart, of the liver, of diabetes, etc. Sarcopenia is an aggravating factor of the liver disease in metabolic steatosis and has been associated with increased fibrogenesis and progression to cirrhosis. Insulin resistance related to metabolic dysfunction and senescence also drives the progression of liver injury in patients with metabolic steatosis. Hence SO is part of a constellation of phenotypical changes associated with metabolic dysfunction and can impact the prognosis as an aggravating factor for liver, metabolic and cardiovascular outcomes.
It is therefore important to diagnose SO but current diagnostic methods are heterogeneous and provide very different figures. We established a new diagnostic method based on a thorough analysis of body composition in a cohort of more than 1400 individuals (overweight or obese). We would like to test this diagnostic method in the large UK Biobank as this collection would allow us to determine whether this diagnosis is related to parameters of muscle strength and function that are otherwise not available when using other datasets and also to understand whether it is associated with other health conditions and their outcome (such as myocardial infarction, cirrhosis, severe diabetes, etc).
If our diagnostic method is confirmed as a valuable tool in this independent group of individuals from the UK Biobank, we will be able to present it to the international scientific community in order to have it validated by other researchers from other countries so that it eventually becomes part of accepted the diagnostic management procedures.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/clinical-correlations-for-genetic-targets-discovered-through-in-vivo-therapeutic-screening

Clinical correlations for genetic targets discovered through in vivo therapeutic screening

Last updated:
ID:
91438
Start date:
21 July 2022
Project status:
Current
Principal investigator:
Dr Martin Borch Jensen
Lead institution:
Gordian Biotechnology, United States of America

In order to develop medicines for human disease, researchers need a way to test new ideas for how to affect the biology of the disease. Even the best systems we use to test effects, treating animals that have the same diseases, have a risk of misleading us because of differences in the biology of animals and humans. This project will supplement extensive animal results in four major diseases, heart fail, lung and liver fibrosis, and osteoarthritis, with data from the human UK biobank cohort in order to ensure that the new medicines we will develop are more likely to work in human patients. This project will continue for at least three years, to be extended for as long as we are getting new information about genes responding to potential medicines.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/clinical-decision-support-for-automated-oct-imagery-interpretation

Clinical Decision Support for Automated OCT Imagery Interpretation

Last updated:
ID:
30043
Start date:
6 April 2018
Project status:
Closed
Principal investigator:
Dr Nicolas Jaccard
Lead institution:
Visulytix Limited, Great Britain

The aim of this research is to quantitatively evaluate the agreement between Human experts and algorithms for the automated detection of pathologies in OCT imagery.

The pathologies investigated include dry & wet age-related macular degeneration (AMD) and diabetic retinopathy. The proposed research aims at significantly improving screening capabilities for prevalent eye pathologies (e.g. age-related macular degeneration). As such, it will enable early detection of said pathologies, greatly improving patient outcome while minimising cost to the providers and healthcare system as a whole. The OCT imagery contained in the biobank uk dataset will be used to develop Artificial Intelligence (AI) algorithms for the detection of eye-related pathologies. This research relies on the availability of large and diverse datasets. Therefore, we would like to request all records of physical eye measurements (including OCT imagery) currently available in the biobank uk dataset.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/clinical-heterogeneity-in-depression

Clinical heterogeneity in depression

Last updated:
ID:
28709
Start date:
1 June 2017
Project status:
Current
Principal investigator:
Professor Jonathan Flint
Lead institution:
University of California, Los Angeles, United States of America

Finding the causes of major depression is a major challenge in global health. One critical question is the extent to which our current classification conceals within the diagnosis of major depressive disorder multiple conditions, each the outcome of different causal pathways. For example there is evidence that the genetic loci that increase risk for depression differ between men and women. We wish to use the phenotypes in Biobank to address whether depression is one condition or many. Depression is the leading cause of morbidity in the developed world and predicted to become the world’s leading cause of morbidity by 2030. It is also a common cause of death: 800,000 people kill themselves every year, and the majority do so in part because they are depressed. Our project to find causes of depression is addressing a global health problem We will classify depression into subtypes, using known or putative causes of heterogeneity, such as age, sex and severity of illness. We will then examine whether the genetic effects on different types of depression are the same. In order to test for clinical heterogeneity we will apply tests that use differences in the way genetic loci act to detect different categories of disease The full cohort will be required for this study


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/clinical-imaging-molecular-phenotyping-and-cardiovascular-outcomes-of-the-cardiovascular-kidney-liver-metabolic-cklm-syndrome

Clinical-imaging-molecular phenotyping and cardiovascular outcomes of the cardiovascular-kidney-liver-metabolic (CKLM) syndrome

Last updated:
ID:
634177
Start date:
3 April 2025
Project status:
Current
Principal investigator:
Dr Nicholas Chew
Lead institution:
National University Hospital (Singapore) PTE. LTD, Singapore

With the recent focus on cardiovascular-kidney-metabolic health, metabolic dysfunction-associated steatotic liver disease (MASLD) remains an under-recognised cardiovascular risk factor. MASLD, chronic kidney disease (CKD), metabolic syndrome and CVD often co-exist, underpinned by similar patho-mechanistic pathways. With the shared risk factors and the crosstalk between organ systems, it is prudent to consider the overlap across these entities, under the broader construct of cardiovascular-kidney-liver-metabolic health (CKLMH). A proposed definition may serve as a starting point in facilitating evaluation of biological and social determinants of CKLMH. In this study, the CKLM syndrome is defined as the presence of CKD, MASLD, and the metabolic syndrome. This study will compare the clinical characteristics, environmental and behavioural factors, imaging data, molecular phenotypes, and cardiovascular outcomes of individuals with CKLM syndrome versus those without.

Objective 1 – To study the association of anthropometrics, physical activity, diet, sleep, cognitive function, myocardial and vascular structure/function, MRI-based body and brain composition, and proteomic/lipidomic profile, in participants with and without CKLM syndrome.

Objective 2 – To identify biomarkers (identified in Aim #1) that are independently associated with cardiovascular disease and mortality, in the population with and without CKLM syndrome.

Objective 3 – To construct a novel risk stratification scoring system for predicting cardiovascular disease and cardiovascular-related mortality using the above biomarkers in participants with and without CKLM syndrome.

This study will be the important next step in characterising the CKLM, identifying independent predictors of MACE, and enhancing conventional cardiovascular risk stratification and intervention targets.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/clinical-importance-of-osteosarcopenia-in-older-people

Clinical Importance of Osteosarcopenia in Older People

Last updated:
ID:
282757
Start date:
23 April 2025
Project status:
Current
Principal investigator:
Dr Nicola Veronese
Lead institution:
King Saud University, Saudi Arabia

Our research project aims to understand a condition called osteosarcopenia, which affects bone and muscle health in older adults. Osteosarcopenia happens when bones become weaker (osteoporosis) and muscles lose strength and mass (sarcopenia) at the same time. We want to learn more about why this happens and how it affects people as they get older. The scientific reason for studying osteosarcopenia is that bones and muscles are closely linked in our bodies. When one weakens, it can affect the other. This condition can make people more likely to fall and get fractures, which can lead to serious health problems, like being less mobile or needing help with daily tasks.
Our project will last for 36 months, during which we’ll study how osteosarcopenia develops and what factors contribute to it. By understanding these factors, we hope to develop better ways to prevent and treat osteosarcopenia in older adults.
The public health impact of our research is significant because osteosarcopenia is a common problem among older adults and can greatly affect their quality of life. By learning more about osteosarcopenia and finding ways to prevent or treat it, we can help older adults stay healthier, more active, and independent for longer. This research could lead to better strategies for promoting bone and muscle health in aging populations, ultimately improving the overall well-being of older adults worldwide.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/clinically-relevant-stratification-through-deep-learning-analysis-of-registry-data-and-extensive-genetic-data-and-identification-of-functionally-relevant-disease-associated-focus-genes

Clinically relevant stratification through deep learning analysis of registry data and extensive genetic data, and identification of functionally relevant disease-associated focus genes

Last updated:
ID:
532555
Start date:
4 February 2025
Project status:
Current
Principal investigator:
Professor Kasper Lage
Lead institution:
Sidera Bio ApS, Denmark

We aim to replicate, validate and build upon findings we have made in Danish data, analysing deep phenotype data (clinical data from the Health Data Authority registries) to create clinically relevant subgroups/clusters. Identifying drivers of these clusters, we will replicate the clustering in the UKBB data and then analyse WGS in the clusters found.
Deep learning methods based on neural networks can capture non-linear correlations and thus represent and identify biologically relevant information. We have developed deep learning models which can stratify patients and identify patterns in disease onset, disease progression, treatment effect, comorbidity, and disease burden. In the Danish data, we have GWAS data for some patients/subgroups. However, the project will be much improved by replication of the clusters in UKBB data.

Pilot case conditions: Cardiometabolic disease, Schizophrenia.
Both are complex diseases with poorly characterized impact of known variants. Better stratification can contribute to earlier diagnosis and better treatment, both directly through professional treatment guidelines and indirectly through a better understanding of the genetics and causal mechanisms, and contribution to the discovery of new drug targets.
The project will utilize data from sources in three countries, the UK (UKBB) Finland and Denmark.

Methods: We use self-supervised learning, where a neural network of Variational Autoencoders (VAEs) is trained to reconstruct the input data. Use of VAEs filters out redundancy and noise while learning higher-level latent features that highlight complex variations among individuals. Cluster analysis of this latent space can be used to stratify groups of individuals with common traits and explore associations between features, such as genomics and health history. We also use transformers, to capture temporality including disease progression.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/clonal-hematopoiesis-and-increased-risk-of-cardiovascular-disease

Clonal Hematopoiesis and increased risk of cardiovascular disease

Last updated:
ID:
103409
Start date:
17 October 2023
Project status:
Current
Principal investigator:
Mr Jiangshan Tan
Lead institution:
Fuwai Hospital Chinese Academy of Medical Sciences, China

Cardiovascular disease is a major global health concern, with atherosclerotic cardiovascular disease being one of the most common types. Despite several recognized risk factors, many patients are diagnosed with arteriosclerosis without any known risk factors, suggesting other factors may be at play. Researchers have discovered a potential new risk factor, known as CHIP (Clonal hematopoiesis of indeterminate potential), which is the presence of an expanded somatic blood-cell clone in persons without other hematologic abnormalities. CHIP has been linked to an increased risk of atherosclerotic cardiovascular disease, with carriers having a 1.9 times higher risk of coronary heart disease and increased artery calcification. As older people are more vulnerable to heart diseases, it is possible that CHIP mutation may be related to the accumulation of time spent with an unhealthy lifestyle. Our project aims to explore the potential relationship between CHIP and cardiovascular diseases, especially in individuals with an unhealthy lifestyle. We plan to collect relevant data from the UK biobank database and analyze it over a period of 36 months. Our findings could lead to more accurate prediction and better management of cardiovascular diseases in the future.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/clonal-hematopoiesis-and-inflammatory-diseases

Clonal hematopoiesis and inflammatory diseases

Last updated:
ID:
101336
Start date:
8 May 2024
Project status:
Current
Principal investigator:
Dr Mikko Myllymaki
Lead institution:
University of Helsinki, Finland

Chronic inflammation is a unifying mechanism behind many aging-associated diseases. Autoimmune diseases such as rheumatoid arthritis (RA) are caused by abnormal immune activation and inflammation, affecting 5% of the population. In contrast, gout is caused by deposition of monosodium urate crystals into joints, resulting in inflammation, joint pain, and functional impairment. Clonal hematopoiesis (CH) refers to competitive growth advantage of a mutated bone marrow hematopoietic stem cell clone in individuals without a diagnosis of an overt hematologic malignancy. CH is common in older individuals and is associated with shorter life expectancy as well as increased cardiovascular disease risk due to enhanced proinflammatory signaling. The convergent inflammatory phenotype warrants studies to understand the clinical connection between CH and inflammation. We hypothesize that CH is common in patients with inflammatory diseases and that CH is associated with higher inflammatory activity in these diseases. In this UKBB project, we will leverage CHIP information from whole exome sequencing and mCA information from SNP arrays to evaluate their association with inflammatory diseases using multivariable models. We will also perform Mendelian randomization to understand whether germline risk factors of inflammatory diseases are also associated with CH risk. The expected project duration is until 2026. Understanding how clonal hematopoiesis contributes to inflammation can elucidate the pathogenesis of inflammatory diseases and can lead to novel therapeutic strategies in future, such as inhibition of the CH clones.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/clonal-hematopoiesis-and-risk-of-solid-cancer

Clonal hematopoiesis and risk of solid cancer

Last updated:
ID:
577088
Start date:
24 January 2025
Project status:
Current
Principal investigator:
Dr Chao Cheng
Lead institution:
Baylor College of Medicine, United States of America

In this application, we propose to leverage the UKBB data to investigate the association between clonal hematopoiesis (CH) and the risk of solid cancers. Recently, we identified a significant association between clonal hematopoiesis mutations (mGMs) and lung cancer risk using data from our in-house cohort (TRICL). Our findings indicated that the presence of CH mutations is significantly linked to an increased risk of lung cancer after adjusting for age, sex, and smoking status. Additionally, we discovered that mosaic chromosomal alterations (mCAs) in white blood cells were associated with an elevated risk of lung cancer, even after accounting for key confounding factors. Notably, we observed a significant increase in ChrX mCAs and mosaic ChrY losses in smokers compared to non-smokers.
In this proposal, we aim to extend our analysis to common solid cancers and investigate the associations of mGMs and mCAs with solid cancers using data from the UK Biobank (UKBB). The UKBB provides comprehensive, high-quality genomic and genetic data for a large number of participants, along with detailed clinical information. We will select individuals diagnosed with solid cancers from the UKBB as the case cohort, focusing on prevalent cancer types such as lung, liver, colorectal, kidney, brain, breast, prostate, uterine, and thyroid cancers. For the control cohort, we will select individuals without any malignancy, matched for clinical factors such as age, sex, and smoking status.
To identify CH mutations in all subjects within the case and control cohorts, we will use whole genome sequencing (WGS) and whole exome sequencing (WXS) data. Additionally, we will leverage high-throughput genotyping arrays and/or WGS data to identify mCAs across the samples. Multivariable regression analyses will be conducted to determine the associations between mGMs and mCAs and the risk of solid cancers. We will also explore variations in these associations across different cancer types. Finally, we w


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/clonal-hematopoiesis-in-the-development-and-progression-of-age-related-diseases

Clonal hematopoiesis in the development and progression of age-related diseases

Last updated:
ID:
150673
Start date:
26 March 2024
Project status:
Current
Principal investigator:
Dr Cecilie Velsoe Maeng
Lead institution:
Copenhagen University Hospital, University of Copenhagen, Denmark

Acquired genetic defects in the blood cells occur in healthy individuals with increased frequency by increasing age. Specific types of these have been associated with increased risk of multiple cancer types, but possibly also other age-related diseases. Only about 1-5% of carriers of these genetic defects develop cancer, while the majority stay healthy. Little is known about reasoning and risk of the occurrence of these genetic defects and what individuals are at higher risk of developing cancer. This study has multiple aims. We intend to examine
– Examine potential risk factors of acquired genetic defects in the blood cells
– Associations between acquired genetic defects, age-related diseases, and mortality

Besides from the data from the UK biobank cohort, we will use data from Danish biobank, databases, and registries. This establishes the opportunity to work with training and validation sets with different demographics to test and strengthen hypothesis and findings.

The study will be performed by Dr. Cecilie Maeng during her PhD-study, which she enrolled September 1st, 2023, at Department of Hematology, Copenhagen University Hospital led by Professor Kirsten Grønbæk.

The perspective is that our study may identify possible early interventions and new drug targets for prevention of development and progression of age-related diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/clonal-hematopoiesis-in-the-development-of-ebv-related-lymphomas

Clonal hematopoiesis in the development of EBV-related lymphomas.

Last updated:
ID:
290648
Start date:
5 November 2024
Project status:
Current
Principal investigator:
Dr Qing-Qing Luo
Lead institution:
The Second Affiliated Hospital of Nanchang University, China

EBV (Epstein-Barr virus) belongs to the herpesvirus family, and nearly 95% of the population will become infected during their lifetime. Although EBV is generally considered a harmless latent virus for most individuals, its reactivation can lead to serious health issues such as lymphoma. The positive rate of EBV is approximately 15% to 30% in patients with lymphoma, though the underlying mechanisms remain unclear. Clone hematopoiesis (CH) represents an expanded somatic cell clone present in individuals without other hematologic abnormalities, and only 0.5% to 1% of CH cases progress to hematologic malignancies annually. To delve deeper into the pathogenesis of EBV-associated lymphomas, we conducted targeted sequencing on 99 cases of EBV-positive lymphomas and uncovered a notable frequency of mutations in CH genes, suggesting a potentially significant role for CH in the development of EBV-related lymphomas. Moreover, previous studies have reported that TET2 and DNMT3A-mediated CH was detected in 60-70% of lymphomas, which is consistent with our findings. Therefore, we propose a scientific hypothesis: CH genes may serve as catalysts, propelling the transformation of EBV-infected individuals towards lymphoma. Our project aims to explore the relationship between CH and EBV-related lymphomas, as well as potential risk factors for CH progression to EBV-related lymphomas. We plan to collect relevant data from the UK Biobank database and analyze it over a period of 36 months. Our research findings may provide important insights for more accurate prediction and better management of EBV-related lymphomas in the future.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/clonal-hematopoiesis-of-indeterminate-potential-and-atherosclerotic-cardiovascular-disease-phenotypes

Clonal Hematopoiesis of Indeterminate Potential and Atherosclerotic Cardiovascular Disease Phenotypes

Last updated:
ID:
62427
Start date:
18 March 2021
Project status:
Current
Principal investigator:
Dr Iftikhar Kullo
Lead institution:
Mayo Clinic, United States of America

Clonal hematopoiesis of indeterminate potential (CHIP) is a relatively common, age-related condition that arises with mutations in an individuals’ blood cells over time. These mutations can occur in a number of different genes. This condition increases the risk for heart attack, although it is unknown if it also increases the risk of related diseases including stroke, peripheral artery disease, and abdominal aortic aneurysm. We suspect that alterations in how the body handles inflammation due to the CHIP mutations are responsible for the increased risk seen for heart attack and related vascular diseases.
Using the genetic data linked with electronic health record data in the UK Biobank and additional databases we plan to:
(1) Identify factors associated with CHIP and assess whether CHIP is predictive of adverse cardiovascular events such as heart attack and stroke.
(2) Investigate whether CHIP is associated with the four major subtypes of vascular disease (heart attack, peripheral artery disease, stroke, and abdominal aortic aneurysm) and with death and adverse events in these patients.
(3) Investigate whether CHIP is associated with markers of inflammation and increased variability in the size of red blood cells.
The expected duration of this project is 36 months including quality control stages for the genetic data, preparing data for analyses, conducting association studies, performing meta-analyses for those traits using UK Biobank and Mayo Biorepository datasets, writing, and publishing the manuscript(s) for this project. The results will increase our understanding of the relationship between CHIP and various cardiovascular diseases and explore the driving factors behind this association including inflammation and abnormal red blood cell generation.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/clonal-hematopoiesis-of-indeterminate-potential-chip-in-aging-and-disease

Clonal hematopoiesis of indeterminate potential (CHIP) in aging and disease

Last updated:
ID:
69235
Start date:
18 June 2021
Project status:
Current
Principal investigator:
Dr Siddhartha Jaiswal
Lead institution:
Stanford University, United States of America

Diseases of aging such as heart disease and stroke are usually thought to occur due to a combination of hereditary and environmental influences. Recently, we discovered that somatic mutations (DNA alterations acquired after birth) in blood cells may be another factor that contributes to these diseases. Approximately 15-20% of people age 70 or older carry a cancer-associated mutation in a substantial proportion of their blood cells, even though the vast majority do not have cancer. This condition has been termed “clonal hematopoiesis of indeterminate potential”, or CHIP. Carriers of CHIP develop blood cancers at a higher rate than the general population, which is expected because it represents the “first-hit” on the path to cancer. Surprisingly, CHIP is also associated with increased all-cause mortality and higher risk of developing non-neoplastic diseases, like atherosclerotic cardiovascular disease. Mechanistically, CHIP mutations increase atherosclerosis due to heightened transcription of inflammatory genes in immune cells. CHIP is the first example of somatic variation acting as a causal factor for common diseases of aging apart from cancer, but a detailed understanding of its consequences for human health is lacking. Because of its link to immune function, inflammation, and aging, we hypothesize that CHIP will also influence many other diseases of aging. Over the next two years, we will use data from UKB to identify novel disease associations of CHIP. We will use novel approaches to interrogate sequence data from these datasets to identify factors that correlate with the rate of growth of mutant cells. We will also leverage the information collected in these biobanks to improve our ability to predict who will suffer adverse consequences, which is a major barrier to developing clinical interventions for CHIP. These studies will broadly advance our knowledge about the causes and consequences of this common, newly described condition of aging.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/clonal-hematopoiesis-of-indeterminate-potential-chip-in-aging-related-diseases-and-inflammation-genetic-and-drug-repurposing-analyses-for-therapeutic-target-prioritization

Clonal Hematopoiesis of Indeterminate Potential (CHIP) in Aging-Related Diseases and Inflammation: Genetic and Drug Repurposing Analyses for Therapeutic Target Prioritization

Last updated:
ID:
89505
Start date:
20 September 2022
Project status:
Current
Principal investigator:
Dr Yap Hang Chan
Lead institution:
University of Hong Kong, Hong Kong

Clonal Hematopoiesis of Indeterminate Potential (CHIP) is the abnormal expansion of mutated blood cell lines without blood disease, and that is detectable as molecular signatures through techniques of genetic sequencing.
CHIP occurs with ageing, and is common. Importantly, CHIP is associated with elevated risks of aging-related diseases such as heart and vascular diseases, stroke, chronic lung disease, abnormal kidney function, as well as inflammatory conditions.
Therefore CHIP represents an important “submerged” clinical iceberg of under-studied health risk that is concealed at the population level. It therefore represents an important underlying cause of morbidities and death risk beyond the conventional risk factors in the ever ageing population.
While inflammation is implicated as an explanatory pathway to explain why CHIP increases the risk of aging-related diseases, the exact underlying scientific mechanisms of CHIP remained poorly understood.
Moreover, currently there is no preventive strategies yet to address the elevated risks of heart diseases and aging-related diseases associated with CHIP.
Therefore, there is a pressing need for further research to unravel the causal mechanistic pathways, and look for factors that can mitigate the disease risks associated with CHIP.

In this proposed research project over 36 months, we will comprehensively dissect the complex relations between CHIP and aging-related diseases through studies of genetic variants related to CHIP and related biological pathways, and studies of drugs and factors that might have the potentials to modulate and ameliorate the heath risks of CHIP. We believe this project will yield important insights in the mechanisms of CHIP leading to heart and aging-related diseases, and shield lights on future prevention strategies and inform therapeutic target prioritization.
The public health impact will be tremendous in terms of allow us to identify factors to abrogate the health risks of CHIP.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/closing-the-loop-towards-precision-medicine-in-obesity

Closing the loop towards precision medicine in obesity

Last updated:
ID:
140043
Start date:
20 March 2024
Project status:
Current
Principal investigator:
Dr Ertunc Erdil
Lead institution:
ETH Zurich, Switzerland

1 – To train strong neural networks using contrastive learning with limited data and annotation on UK Biobank imaging data to identify glucocorticoid (GC) dependent pathway activation and fine-tune them for various tasks.
2 – To train neural networks on UK Biobank data to predict some health indicators and evaluate the model’s performance on the consortium’s in-house datasets. Furthermore, investigate ways for multi-task learning using UK Biobank and in-house datasets.
3 – To learn representations that are useful for both segmentation and prediction of metabolic health indicators.
4 – To apply the neural networks trained on the consortium’s in-house data for GC pathway activation prediction to the UK Biobank dataset and investigate the correlation between the predicted activation and some metabolic health indicators already available in UK Biobank. Ultimately, these predictions can be added to the UK Biobank data.
5 – To investigate the correlation between GC pathway activation and brain structures.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cluster-analysis-of-genetic-clinical-environmental-and-lifestyle-exposures-in-relation-to-breast-cancer-risk-and-progression

Cluster Analysis of Genetic, Clinical, Environmental, and Lifestyle Exposures in Relation to Breast Cancer Risk and Progression

Last updated:
ID:
947018
Start date:
4 November 2025
Project status:
Current
Principal investigator:
Dr Zhu Zhu
Lead institution:
Southeast University, China

This project aims to investigate how diverse exposures jointly influence breast cancer risk and progression by identifying clusters of co-occurring risk factors.
Research questions:
1.What distinct multi-dimensional exposure patterns exist among women (including genetic, clinical, lifestyle, environmental, sleep, and dietary factors)?
2.How are these exposure clusters associated with breast cancer incidence?
3.Do these clusters predict breast cancer progression outcomes such as metastasis or complications?
Objectives:
1.Identify clusters of women sharing similar profiles across genetic risk, clinical characteristics, environmental exposures (e.g., air pollution), lifestyle behaviours (smoking, alcohol use, physical activity), sleep traits, and dietary intake.
2.Examine associations between these exposure clusters and breast cancer incidence.
3.Assess whether these clusters are linked to breast cancer progression outcomes (e.g., metastasis, complications) using cancer registry and hospital episode data.
Scientific rationale:
Breast cancer risk is shaped by multiple interacting factors that rarely occur in isolation. While individual risk factors have been well described, less is known about how they cluster in real-world populations and jointly influence disease development and progression. Moreover, integrated analyses combining genetic susceptibility (e.g., polygenic risk scores), clinical risk factors, sleep, diet, and environmental exposures are rare.
Using UK Biobank’s large, deeply phenotyped cohort with genetic data, lifestyle assessments, environmental measures, and cancer registry linkage will enable robust identification of meaningful exposure patterns. Understanding these clusters can support risk stratification, inform targeted prevention strategies, and improve insights into breast cancer etiology and prognosis.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/clustering-of-cardiometabolic-risk-factors-and-their-association-with-physical-activity-and-sedentary-time-in-women-with-and-without-polycystic-ovary-syndrome-a-principal-component-analysis

Clustering of cardiometabolic risk factors and their association with physical activity and sedentary time in women with and without polycystic ovary syndrome: a principal component analysis.

Last updated:
ID:
52771
Start date:
16 October 2019
Project status:
Closed
Principal investigator:
Dr Chris Kite
Lead institution:
Aston University, Great Britain

Polycystic ovary syndrome (PCOS) affects many women worldwide and causes a range of symptoms such as acne, excess hair, irregular periods, and sometimes, infertility. Women with PCOS are also more likely to be obese, have high blood pressure and high cholesterol which can lead to diabetes and heart disease.

Exercise is often recommended to manage the symptoms of PCOS. However, less is known about the effects of day-to-day activities. Day-to-day activities are the things done for work, walking from place to place, household chores or sports that are played for fun. We are also interested in the time spent sitting. Those who sit for long periods of time have a higher risk of disease, and when activity levels are low, the risk is even greater. To our knowledge, this has not been investigated in women with PCOS.

Participants in the UK Biobank have reported their physical activity and sitting behaviours. We will assess these reported values to find out whether being more physically active, and sitting less reduces the risk of developing further conditions in PCOS. We will also compare this data to a group of women without PCOS to find out whether it is more (or less) important if you have been diagnosed with PCOS.

Gaining a better understanding about the role of being active, and high sitting time in PCOS will help to develop treatment guidelines and shape the information provided by health professionals. It will also help to educate women with PCOS about the physical and mental benefits of becoming more active.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/co-analysis-of-the-international-mouse-phenotyping-consortium-impc-data-and-the-uk-biobank-exome-sequencing-and-blood-screen-data-to-identify-gene-phenotype-associations

Co-analysis of the International Mouse Phenotyping Consortium (IMPC) data and the UK Biobank exome sequencing and blood screen data to identify gene-phenotype associations

Last updated:
ID:
62659
Start date:
11 January 2021
Project status:
Closed
Principal investigator:
Dr Violeta Munoz-Fuentes
Lead institution:
European Bioinformatics Institute (EBI), Great Britain

The UK Biobank and the International Mouse Phenotyping Consortium (IMPC) attempt to identify associations between genes and traits, that is, gene-phenotype associations. While the UK Biobank relies on genetic and phenotypic data collected for a large number of participants, the IMPC relies on phenotypes collected for mouse knockouts, that is, mice in which one gene has been knocked out (inactivated or “switched off”).

Notably, an overlap in procedures to describe blood traits exists between the UK Biobank and the IMPC, involving 39 blood phenotype parameters, including white and red cell counts and biochemical data. Taking advantage of this overlap, this project will focus on associations between genes and blood traits. Our aim is to identify human genes contributing to blood molecular phenotypes by integrating IMPC and UK Biobank gene-phenotype associations.

Blood trait data has been collected for all UK Biobank participants. In terms of genetic data, in this study we plan to focus on whole-exome sequence (WES) data that has now been generated for 150,000 participants of the UK Biobank cohort. To establish associations between blood phenotypes and human genetic variants, we plan to use so-called gene-based burden methods. These methods can be used to highlight genetic variants that are likely contributing to a phenotype (or trait).

This project will highlight variants likely contributing to abnormal blood phenotypes associated with disease, and also constitutes an interesting opportunity to relate alterations in blood physiology, the immune system, enzymatic activity and blood metabolism between human and mouse, and give insight into the suitability of these mouse lines informing about human disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/co-morbidities-and-risk-factors-of-inflammatory-rheumatological-conditions

Co-morbidities and risk factors of inflammatory rheumatological conditions

Last updated:
ID:
7996
Start date:
1 January 2015
Project status:
Closed
Principal investigator:
Dr Suzanne Verstappen
Lead institution:
University of Manchester, Great Britain

Chronic inflammatory rheumatological conditions such as rheumatoid arthritis (RA) and psoriatic arthritis (PsA) and their co-morbidities cause long-term disability. Both environmental and genetic risk factors contribute to disease susceptibility but they have seldom been studied in the same population. A comprehensive study in a large population is therefore required.

Aims:
1. To compare rates and consequences of co-morbidities in patients with inflammatory rheumatological conditions with those without the condition.
2. To develop predictive models to identify individuals in the general population at high risk of developing RA, and psoriasis patients at high risk of developing PsA. Compared to the general population patients with RA or PsA have increased rates of co-morbidities such as cardiovascular diseases, respiratory diseases, cancer and depression, which in turn may lead to a lower quality of life and decreased physical functioning. Understanding the risk factors influencing the development of these rheumatological conditions, and their associated co-morbidities, will allow early clinical intervention to reduce disability and result in improved outcomes for patients. This study proposal meets BioBank?s stated purpose as it aims to prevent illness through early diagnosis resulting in improved health in the population. First we will compare rates of cardiovascular and respiratory disease and cancer between groups of patients with different self-reported inflammatory rheumatological conditions (e.g. RA, PsA and Lupus) to rates in the rest of the UK BioBank. We will also evaluate the impact of these co-morbidities on physical functioning. Secondly, we will attempt to validate known risk factors in addition to identifying novel factors that contribute to disease. All identified risk factors will be used to create a prediction model. The model?s performance will be tested in predicting incident cases between the first and second visit. This study requires access to data from the full UK Biobank cohort, including follow-up data on the subset who attended for a second visit and primary care data, in order to identify the various prevalent and incident cases of disease and to use the remainder of the cohort as a control population. In addition we would like to request access to the genotype data when it is available in 2015 and also the primary care data once available, to help identify incident cases of disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/coffee-and-the-risk-of-chronic-diseases-including-liver-cancer-liver-cirrhosis-type-2-diabetes-mellitus-and-chronic-kidney-disease

Coffee and the risk of chronic diseases, including liver cancer, liver cirrhosis, type 2 diabetes mellitus and chronic kidney disease.

Last updated:
ID:
26877
Start date:
18 June 2019
Project status:
Current
Principal investigator:
Dr Ryan Malcolm Buchanan
Lead institution:
University of Southampton, Great Britain

Observational studies indicate that coffee consumption may protect against certain chronic diseases, including liver cancer, cirrhosis, type 2 diabetes mellitus and chronic kidney disease. However, it is uncertain whether there is an underlying causal mechanism or if the observations result from confounding factors. Our aim is to investigate whether an association between the aforementioned chronic diseases and coffee consumption exists among UK Biobank participants, and to test the causality of the associations using genetic analysis. The aim of this work is to support the development of new strategies for the prevention of chronic disease, including liver cancer, cirrhosis, type 2 diabetes mellitus and kidney disease. We will apply epidemiological and genetic methods to the data in Biobank. We will first determine if there are associations between baseline coffee consumption and outcomes relating to liver, kidney and metabolic diseases. We will then test for effect modification of those associations by risk factors (e.g. smoking, BMI) and genotypes related to coffee metabolism. Finally, we will assess the causality of the associations by performing a Mendelian randomisation analysis, which will involve performing a genome wide association study to identify for genetic markers of coffee consumption. Full cohort


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cognition-across-the-lifespan

Cognition across the lifespan

Last updated:
ID:
96012
Start date:
11 January 2023
Project status:
Current
Principal investigator:
Professor Marvin Chun
Lead institution:
Yale University, United States of America

Human functional magnetic resonance imaging (fMRI) research has now advanced to quantify the psychological traits and behaviors of individuals from their brain scans. Such personalized, precision fMRI, validated with big data sets, is essential for neuroimaging to impact clinical practice and public health. As a promising example to advance personalized fMRI’s potential, we developed a brain-based general attention measure to quantify a person’s attentional function from their fMRI scans. Attention has a ubiquitous role in perception and cognition, and attention deficits are common in mental illness and as symptomatic of brain damage. The goal of this proposal is to expand the brain-based general attention measure to the UK Biobank dataset. We will further validate and improve our models to measure cognition, study the effects of aging, and test the impact of health challenges such as Covid-19. As a long-term goal, our methods and models can advance the development of personalized fMRI, useful for behavioral assessment, with potential for clinical applications to quantify cognitive deficits such as ADHD, dementia, schizophrenia, and trauma from brain scans alone, like one would use a thermometer to measure fever or a monitor to measure blood pressure. Brain-based biomarkers can facilitate diagnosis, treatment, and prevention.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cognitive-and-chronobiological-markers-of-cancer-survival

Cognitive and chronobiological markers of cancer survival

Last updated:
ID:
53734
Start date:
6 December 2019
Project status:
Current
Principal investigator:
Ms Erin Taniyo Kaseda
Lead institution:
Rosalind Franklin University of Medicine and Science, United States of America

As cancer treatment continues to improve, there will be an estimated 70 million cancer survivors worldwide by the year 2020. At the beginning of cancer treatment, cognition has been associated with cancer survival, such that higher cognition has been linked to better survival. Additionally, chronobiological markers have previously been implicated in health outcomes, such that chronic sleep and circadian rhythm disruption may be risk factors for developing cancer and a variety of neurocognitive disorders. However, there has been limited research on the connection between premorbid cognition, sleep and circadian rhythm robustness, and cancer survival. The primary objective of this investigation is to evaluate whether premorbid cognition and chronobiological disruption are predictive of survival among patients diagnosed with cancer. The specific aims of the proposed research are: (1) to investigate the relationship between premorbid cognition, sleep, and circadian rhythm and likelihood of cancer diagnosis; (2) to assess the degree to which cognition impacts survival after cancer diagnosis; and (3) to investigate sleep and circadian rhythm disruption as a predictor of survival and to examine whether this association between cognition and survival is explained by a mediator – circadian rhythm disruption. The present study was designed to utilize the approximately 500,000 data points in the UK Biobank. This project is expected to take approximately 12 months to complete. Findings have excellent potential to help clinicians establish risk profiles for cancer patients, develop more effective treatments, better monitor ongoing treatment to help patients and families plan for likely outcomes, and better target treatment to patients based on cognitive, and sleep/circadian biomarkers.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cognitive-auditory-and-other-health-outcomes-after-subarachnoid-haemorrhage-and-genetic-determinants

Cognitive, auditory and other health outcomes after subarachnoid haemorrhage, and genetic determinants

Last updated:
ID:
49305
Start date:
8 August 2019
Project status:
Closed
Principal investigator:
Professor Ian Galea
Lead institution:
University of Southampton, Great Britain

Subarachnoid haemorrhage is a common cause of stroke in young and middle-aged adults caused by a bleed on the brain. It occurs after rupture of a swelling (or aneurysm) on a major artery supplying the brain and results in release of blood over the brain surface (or the subarachnoid space), which then clots. In survivors, subarachnoid haemorrhage results in substantial loss of quality of life, and a significant cost to the UK economy due to inability to return to work. There is increasing recognition that, although people with a history of SAH look outwardly healthy, they have substantial “hidden” disability which impairs their daily functioning. These hidden deficits mostly consist of problems with memory, concentration and processing of heard information such as speech and music. There are no treatments to prevent or improve recovery from these deficits. Hence a better understanding as to what contributes to a poor outcome after brain haemorrhage is needed. There are over a thousand UK BioBank participants with a history of subarachnoid haemorrhage. First we intend to confirm that employment, physical activity, memory and concentration and hearing is reduced in people with a history of subarachnoid haemorrhage compared to people who have not had a subarachnoid haemorrhage. Then we intend to see whether any specific variation in the genetic code of individuals with a history of subarachnoid haemorrhage predisposed them to a worse or better outcome. It is possible that certain genetic variations may reflect a biological process which can be improved or inhibited with available drugs. We hope that this study will demonstrate the importance of the hidden deficits after subarachnoid haemorrhage, identify potential new treatments and ultimately help improve the management of people with subarachnoid haemorrhage. Finally this study will provide two students, working under our close supervision, with experience in research.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cognitive-factors-in-speech-in-noise-perception

Cognitive Factors in Speech-in-Noise Perception

Last updated:
ID:
611415
Start date:
10 April 2025
Project status:
Current
Principal investigator:
Professor Ingrid Suzanne Johnsrude
Lead institution:
University of Western Ontario, Canada

This project investigates whether a general cognitive factor better explains speech-in-noise (SiN) intelligibility than individual cognitive task scores. While the role of cognition in SiN perception is widely accepted, the contributions of different domains are unclear.

Working memory (WM), typically assessed by the Reading Span (RSPAN) task, consistently correlates with SiN (Akeroyd, 2008), but this does not imply that WM is uniquely critical for SiN tasks. Instead, a broader cognitive factor may influence both RSPAN and SiN performance. Cognitive abilities often intercorrelate, forming the ‘positive manifold’ (Spearman, 1904; 1927), which suggests that diverse cognitive tasks may rely on a shared set of resources, summarized by a common factor ‘g.’ Consequently, the association between WM and SiN may reflect this broader cognitive factor rather than a specific reliance on WM alone. Using data from the UK Biobank, we will conduct factor analysis to determine how SiN performance on the Digit Triplet Test aligns with cognitive factors beyond single-task associations. We expect a common factor, consistent with ‘g’, to emerge, potentially alongside other factors reflecting other clusters of tests. This study aims to clarify the cognitive dimensions underlying SiN perception, ultimately contributing to a more comprehensive model of the cognitive demands involved in effortful listening.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cognitive-function-physical-activity-and-age-analysis-of-the-uk-biobank

Cognitive function, physical activity and age: analysis of the UK Biobank

Last updated:
ID:
10813
Start date:
2 November 2015
Project status:
Closed
Principal investigator:
Professor Thomas Yates
Lead institution:
University of Leicester, Great Britain

We main to investigate the association of daily physical activity time (walking, moderate, vigorous) and sedentary time (computer use, driving and TV viewing) on indices of cognitive function throughout different ages in adulthood in order to identify where (at what age) associations between physical activity and age is strongest and what physical activities are likely to maximise the effect. Declining cognitive function is an inevitable consequence of aging. However, research has shown that degree of cognitive impairment is associated with, or improved by, the amount of moderate-to-vigorous physical activity undertaken in older adults. Physical activity has also been associated with cognitive function in younger and middle-aged adults. However, previous research has not adequately quantified the association between different types of physical activity, sedentary behaviours, cognitive function and age. This proposal therefore seeks to address this need in order to provide insight into how cognitive decline may be targeted in the future. We will quantify the extent to which different types of physical activity and sedentary behaviour are associated with measures of cognitive function and whether these associations vary across different age groups. This will enable assessment of what physical activities are most strongly associated with cognitive function and how the strength of association varies with age. This analysis will use the full cohort where possible. Included cogntive function variables will be analysed on a pairwise basis


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cognitive-networks-in-the-brain-variability-within-the-population-and-predictive-power-for-disease

Cognitive networks in the brain: variability within the population and predictive power for disease

Last updated:
ID:
162321
Start date:
4 April 2024
Project status:
Current
Principal investigator:
Dr Amaia Carrión Castillo
Lead institution:
Asociación Basque Center on Cognition Brain and Language, Spain

Multiple developmental, psychiatric and neurological conditions (e.g. e.g. dyslexia, intellectual disabilities, schizophrenia, aphasias, dementias) manifest through below-average performance in tasks that tap into different aspects of cognitive functions such as language, memory or attention. These cognitive functions enable individuals to fulfil their full potential and encompass an important dimension for a person’s well-being. Thus, characterizing the underlying networks in the brain, and understanding environmental and genetic factors affecting individual variability in these traits has the potential to provide useful risk predictors.
We will first investigate cognitive networks in the imaging subset of the UK Biobank, to characterize how these neurobiological correlates behave in the healthy and atypical brains. To this aim, we will assess structural (grey and white matter) and functional (resting state) MRI measures. We will assess the cortical reorganization that occurs in patients, and whether it has links with the aging processes that occur even in the absence of disease.
Next, we will perform hypothesis-free analyses of relevant brain networks to see whether they are associated with environmental or genetic variables. This will result in the identification of potential environmental risk factors, as well as genome-wide association results. Furthermore, we will perform downstream genetic analyses to further characterize the effects of these brain regions at the genetic level: heritability, genetic correlation, polygenic score analyses.
The validity of these brain correlates as useful proxies to characterize the cognitive networks will be tested through a multi-level analysis: assessing the relationship with the available cognitive tasks in the UK Biobank, and by means of genetic correlations/polygenic score analyses with other datasets.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cognitive-neuroimaging-genomics-to-advance-our-understanding-of-brain-function-and-its-disorders

Cognitive neuroimaging genomics to advance our understanding of brain function and its disorders

Last updated:
ID:
75807
Start date:
8 December 2021
Project status:
Current
Principal investigator:
Dr Xiangzhen Kong
Lead institution:
Zhejiang University, China

Aims: How do genetic and early life factors regulate brain circuits that underlie cognition (e.g., spatial navigation, memory, and language) in healthy aging and diseases (e.g., Alzheimer’s disease)? This project aims to explore this question in humans, by combining various types of measures of individuals in a large-scale sample.
Rationale: In the proposed project, we plan to explicitly incorporate prior knowledge of brain networks underlying various cognitive functions (e.g., spatial navigation, memory, and language) into the integrative analysis of various data including cognition, brain imaging, early life experience, and genetic data. Genetic and life experience variables will be associated with functional networks in the human brain. The top genetic variants and early life factors revealed could provide candidates for further cognition and brain studies in healthy aging and diseases.
Project duration: The project will take 36 months.
Public health impact: The results would reveal new genetic variants and environmental factors associated with human cognition and brain networks in healthy aging and disease. We expect that this knowledge will be greatly helpful for future public health policy on diagnosis and treatment of diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cognitive-outcomes-in-breast-cancer-survivors

Cognitive outcomes in breast cancer survivors

Last updated:
ID:
94496
Start date:
26 April 2023
Project status:
Current
Principal investigator:
Dr Kathleen Van Dyk
Lead institution:
University of California, Los Angeles, United States of America

The purpose of this project is to better understand whether breast cancer treatment affects cognition and risk for dementia in aging women. Some effects of breast cancer and its treatment may result in accelerated brain aging, and could put some women at increased risk for developing cognitive impairment earlier in life. Most studies to date have been too small to fully understand if women who may be already at risk for dementia (e.g., if they carry the APOE4 allele that increases risk for Alzheimer’s disease) might be at greater risk following treatment for breast cancer. This proposal aims to examine the UK Biobank longitudinal data to address: a) whether breast cancer and its treatment affect a woman’s risk for cognitive impairment and dementia; and b) which women with a history of breast cancer may be the at highest risk. The project duration is approximately 3-5 years and the potential public health impact includes improving identification of risk factors for cognitive decline and dementia in the large percentage of women affected by breast cancer.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cognitive-outcomes-in-people-with-behavioural-and-brain-disorders-within-uk-biobank

Cognitive outcomes in people with behavioural and brain disorders within UK Biobank

Last updated:
ID:
11332
Start date:
1 February 2015
Project status:
Closed
Principal investigator:
Dr Breda Cullen
Lead institution:
University of Glasgow, Great Britain

The aim of this research is to improve our understanding of variation in cognitive performance in adults with behavioural and brain disorders such as depression, bipolar disorder and multiple sclerosis. We will investigate the nature and extent of cognitive impairment in these groups compared to healthy controls, and we will develop multivariate models to explore the relationship between cognitive performance and medical status, demographic and lifestyle factors, and genetic markers. Cognitive impairment is common and functionally disabling in patients with behavioural and brain disorders, but it remains poorly understood at an individual level. This cross-sectional research will contribute to the refinement of hypotheses regarding risk factors for cognitive impairment, providing a foundation for future longitudinal research focused on understanding, preventing and treating cognitive impairment in these groups. This research will comprise a series of cross-sectional studies of baseline cognitive data from the UK Biobank resource. Complex statistical models will be used to estimate the relationship between key risk factors (e.g. medical status and genetic markers) and cognitive performance, while taking into account the additional influence of other demographic, social and lifestyle factors. This research will make use of the full UK Biobank cohort. Sub-groups with behavioural and brain disorders will be identified, and the remainder of the cohort will serve as a control group for comparison.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cognitive-reserve-markers-and-dementia-incidence

Cognitive reserve markers and dementia incidence

Last updated:
ID:
56580
Start date:
22 September 2020
Project status:
Current
Principal investigator:
Professor Esme Fuller-Thomson
Lead institution:
University of Toronto, Canada

Aims
It is not clear exactly what activities contribute to an increased ability to resist brain damage. It is believed that educational attainment, work complexity and mentally stimulating leisure activities can enhance cognitive reserve, but more studies are needed to achieve conclusive results. Therefore, this project aims to investigate what life-course activities individually increase the amount of cognitive reserve. This project also aims to combine these various markers into one cognitive reserve score, and test if increased levels protect against dementia.
Scientific rationale
The brain has a natural capability to repair itself and resist neuropathological damage, but this ability is severely affected by dementia progression. Over the years, scientists have been puzzled by marked differences between individuals who are able to resist and recuperate from brain damage and those who are not. Hence, the ongoing research question is: What makes some people cope with brain damage and neurodegeneration better than others?
Around 30% of autopsy reports show that older individuals who preserved their mental abilities, show damage in their brains after they pass away. To explain the inconsistency between brain damage and its manifestation, it has been proposed that the combined knowledge and experiences individuals acquire through their life protect the brain by increasing its ability to resist damage. The enhanced protection that results from activities carried out throughout the lifetime has been named cognitive reserve. To date, several studies have found evidence supporting that high levels of cognitive reserve can protect against dementia.
Project duration
5 year duration, starting January 2020 up to Dec 2025
Public health impact
Individuals with high cognitive reserve can delay or fully avoid the effects of dementia; even if they develop the disease, they are able to overcome its symptoms (memory loss, disorientation, language problems, poor judgment, and mood changes) and continue living an independent lifestyle. Hence, promoting an enhanced cognitive reserve throughout the lifetime will benefit the lives of people who will hopefully avert neurological conditions such as dementia.
This project will also investigate the extent to which cognitive reserve can continue to enrich cognitive performance through lifestyle activities, even after a dementia diagnosis. People already affected by dementia symptoms can potentially slow their decline by participating in cognitive reserve enhancing activities.
Based on the results of this study, cognitive reserve enhancing policies and programs can be put into practice in the UK, extending the quality of life of millions of people and their families in the country.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cognitive-traits-and-brain-mechanisms-across-insomnia-related-mental-health-complaints

Cognitive traits and brain mechanisms across insomnia-related mental health complaints.

Last updated:
ID:
171264
Start date:
13 February 2025
Project status:
Current
Principal investigator:
Professor Eus J W Van Someren
Lead institution:
Netherlands Institute for Neuroscience, Netherlands

Mental health issues caused by poor sleep are widespread and often occur together, accounting for a major proportion of the global mental health burden. The primary aim of our research is to understand these sleep-related mental health conditions better, especially insomnia, anxiety, depression, and stress-related disorders. Specifically, we are interested in understanding their interactions and common factors, like being overly alert (hyperarousal), that contribute to these disorders.

Right now, the treatments available only work moderately well, and many people continue to experience symptoms even after treatment. We think that if we can identify the brain correlates tied to these disorders, we can come up with better treatments. For example, we hypothesize that there might be common symptoms shared by different mental health issues, possibly related to genetics and brain deviations that could be targets for transdiagnostic treatments.

Over the next three years, we plan to get a clearer picture of these sleep-related disorders. We want to see how they relate to each other and identify any shared symptoms. Once we know more about these, we will study the brain’s structure and functions to understand their role in these disorders. This could help us see the shared symptoms that connect different mental health issues and how the brain plays a part in it.

We hope our findings will help doctors and researchers understand the connection between poor sleep and mental health problems better and that this will help to improve treatment. Furthermore, the cross-diagnostic symptoms we aim to pinpoint will serve as a valuable foundation for further studies in sleep-related disorders.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cohort-study-of-risk-factors-of-acute-and-chronic-cardiovascular-events-in-covid-19-patients

Cohort study of risk factors of acute and chronic cardiovascular events in COVID-19 patients

Last updated:
ID:
70844
Start date:
22 February 2021
Project status:
Closed
Principal investigator:
Professor Martin Gulliford
Lead institution:
King's College London, Great Britain

This research aims to inform patient management during the present COVID-19 coronavirus pandemic. Early findings suggest that patients with COVID-19 commonly experience conditions affecting the heart and blood vessels (cardiovascular conditions) such as heart failure. Acute heart failure often leads to very severe outcomes including death. We are also interested in understanding the risk factors for longer-term cardiovascular outcomes in COVID-19 patients such as chronic heart failure. Those with cardiovascular risk factors, such as high blood pressure, and who have experienced cardiovascular conditions in the past, seem to be at increased risk of poor COVID-19 outcomes. However, studies have reported several cases of cardiovascular events following a COVID-19 diagnosis in patients without a history of cardiovascular disease. It appears that the SARS-CoV-2 virus that causes COVID-19, might be capable of directly causing damage to the heart and more research is required to understand which patients are likely to benefit from dedicated, specialist cardiac care and monitoring. This study will use UK Biobank data to collate cardiovascular risk factors in COVID-19 patients to assess their association with cardiovascular events including acute heart failure, chronic heart failure and death. We will analyse data from patients who have diagnoses of heart failure or other cardiovascular conditions, comparing biomarkers, risk factors and disease progression among those with a preceding COVID-19 diagnosis to those without.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/combination-of-polygenic-risk-score-and-classical-risk-factors-to-predict-pan-cancer-risks

Combination of polygenic risk score and classical risk factors to predict Pan-cancer risks

Last updated:
ID:
94939
Start date:
10 November 2022
Project status:
Current
Principal investigator:
Professor Xiaoping Miao
Lead institution:
Wuhan University, China

Our project aims to uncover novel, modifiable environmental risk factors in pan-cancer based on two-stage population studies. With the advance in understanding cancer risk factors, translating lifestyle, environmental, and genetic risk factor information into actionable clinical information is the next step in developing personalized prevention. Therefore, we will incorporate these environmental lifestyle factors, with the known common genetic variants to develop risk prediction models using machine learning and deep learning methods. We will further expand the risk prediction analysis to define the optimal starting age for screening. These models may be useful to prioritize those at high risk for targeted prevention or intervention and to reduce emphasis on those at low risk of developing cancers, thereby optimizing utilization of screening in clinical practice with individually tailored prevention strategies.

The project is scheduled to begin in October 2022 and be completed in October 2025. The research plan is as follows:
1)October 2022-April 2023: data application and processing.
2)May 2023-December 2024: (1) identifying novel risk factors in pan-cancer; (2) building a risk prediction model; (3) evaluating risk prediction models.
3)January 2025-October 2025: summarizing research results and writing research papers.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/combined-analysis-of-multi-source-heterogeneous-data-for-depression-detection-and-diagnosis

Combined analysis of multi-source heterogeneous data for depression detection and diagnosis

Last updated:
ID:
87530
Start date:
24 May 2022
Project status:
Current
Principal investigator:
Professor Zhongchun Liu
Lead institution:
Wuhan University, China

More than 350 million people in the world suffer from depression, and depression has become the fourth largest disease in the world. It is predicted that depression will become the first disease in the global disease burden in 2030. However, diagnostic biomarkers for depression are lacking, and previous studies on depression have reported many inconsistent results, which may be attributed to the small sample size and within-sample demographic and clinical heterogeneity. Using the UK Biobank database, this study aims to integrate clinical data, psychosocial factors, neuroimaging, and genetic data of depression, and use large sample data to explore precise objective biomarkers related to depression.
Combining multiple computer methods such as machine learning and deep learning, and integrating genetic, brain imaging and cognitive/emotional data, and demographic data from the UK Biobank, the onset of depression is predicted through cross-sectional and prospective follow-up data. We will examine associations between genes, brain imaging and cognitive/mood data, demographic data, and explore how these associations are mediated by brain MRI or mediated by biomarker variables (inflammatory markers, vitamins, etc.) of. It is expected to screen out 2-3 accurate, sensitive and effective monitoring biomarkers for depression, and build a prevention and treatment network for depression research. Provide active and effective scientific and technological support for accelerating technological breakthroughs in the prevention and control of depression, controlling the growth of medical expenses, promoting the rational and standardized application of technology, reducing medical and social burdens, and curbing the high incidence and mortality of major chronic diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/combined-associations-of-body-weight-and-lifestyle-factors-with-arterial-health-cardiovascular-events-and-mortality

Combined associations of body weight and lifestyle factors with arterial health, cardiovascular events and mortality

Last updated:
ID:
62594
Start date:
6 October 2020
Project status:
Current
Principal investigator:
Professor Luigi Fontana
Lead institution:
University of Sydney, Australia

Western diets, which typically contain large amounts of animal and energy-dense foods, together with a sedentary lifestyle and smoke, impair vascular health and drive cardio- and cerebro-vascular disease. Research from our team has demonstrated the numerous benefits of lifestyle programmes to improve health, however there are still a number of questions to be answered and the UK Biobank provides a unique opportunity to investigate them.

Some of the benefits of this dataset is its large size (>500,000 adults), its depth analysis of dietary behaviours, and the biggest dataset of objective physical activity. This dietary and physical activity data, along with information on sleep, smoking and alcohol behaviour will be combined to create a Healthy Lifestyle Score, which has previously been done by members of team by using the Harvard Health Professionals Follow-up Study (Veronese et al. 2016). Using this score, we aim to investigate the following questions:
1) What is the relationship between the Healthy Lifestyle Score and arterial health. The reason we want to measure this is because arterial health is one of the first signs that an individual is developing cardiovascular disease (CVD) and stroke.
2) How does the Healthy Lifestyle Score predict CVD onset, CVD complications and CVD death. The UK Biobank gives researchers access to individual medical records which means we can measure which lifestyle behaviours have an impact upon CVD onset and death.
3) How does body weight affect the relationship between Healthy Lifestyle Score and CVD. Some research suggests that being overweight or even obese can protect you from some chronic disease and premature mortality. We will be investigating whether taking into account healthy lifestyle behaviours changes this relationship.
4) The impact of diet upon CVD. Dietary behaviour will be explored in more depth as individuals in the UK Biobank study were asked about their diet behaviour 4 times over 1 year. With this data we can explore what the impact of different micro- and macro-nutrients upon CVD risk and death.

Currently, CVD is the largest cause of disease and death worldwide and a lot of energy and money is spent around managing and treating this condition. Data from this project will highlight whether efforts should be directed towards preventative strategies such as encouraging and prescribing healthy lifestyle behaviours. This project will take 2 years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/combined-effect-of-the-genetic-and-lifestyle-determinants-of-metabolic-syndrome-on-cardiometabolic-risk

Combined effect of the genetic and lifestyle determinants of metabolic syndrome on cardiometabolic risk

Last updated:
ID:
32683
Start date:
26 October 2018
Project status:
Current
Principal investigator:
Professor Torben Hansen
Lead institution:
University of Copenhagen, Denmark

Metabolic syndrome is a key risk factor for the development of type 2 diabetes and cardiovascular disease. This proposal seeks to leverage the large sample size of the UK Biobank data to study i) the effect of known genetic loci associated with metabolic syndrome or its subcomponents with the risk of cardiometabolic disease; ii) to examine whether these genetic loci interact with modifiable lifestyle risk factors of cardiometabolic disease; iii) to test whether the loci are associated with the levels of modifiable lifestyle risk factors; and iv) to estimate genetic correlations between subcomponents of the metabolic syndrome and its comorbidities By providing novel and clinically relevant information on the combined impact of the genetic and lifestyle determinants of metabolic syndrome and cardiometabolic risk, the proposed research is congruent with the stated aim of the UK Biobank to improve ?the prevention, diagnosis, and treatment of a wide range of serious and life-threatening illnesses?. The research will be undertaken by conducting epidemiological analyses to study associations between the genetic and lifestyle determinants of metabolic syndrome with cardiometabolic disease risk. Additional analyses will be performed to examine synergistic effects between the genetic and lifestyle risk factors, and to study the overall genetic relationships between subcomponents of the metabolic syndrome and its comorbidities We would like to include the full cohort


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/combined-effects-of-genetic-and-non-genetic-factors-on-digestive-system-diseases-and-risk-prediction-under-different-metabolic-states

Combined Effects of Genetic and Non-Genetic Factors on Digestive System Diseases and Risk Prediction under Different Metabolic States

Last updated:
ID:
354686
Start date:
14 November 2024
Project status:
Current
Principal investigator:
Dr Qinxing Cao
Lead institution:
University of Electronic Science and Technology of China, China

Objective: This study aims to explore the combined effects and risk prediction of genetic and non-genetic factors on digestive system diseases under different metabolic states, and to establish predictive models to help identify risk factors at the population level for early intervention.
Scientific Basis: Digestive system diseases, including gastroenteritis, peptic ulcer, fatty liver disease, stomach cancer, colon cancer, and liver cancer, severely affect the health of the global population. These diseases not only affect patients’ quality of life but also impose a heavy economic burden on the global healthcare system. Although existing studies have pointed out the important roles of genetic, environmental, lifestyle factors, and metabolic state in the occurrence and development of these diseases, there is currently a lack of prospective studies to systematically evaluate the combined effects of genetic and non-genetic factors on digestive system diseases under different metabolic states. Through large-scale analysis of prospective data, we can effectively identify risk factors for digestive system diseases, thereby enabling early intervention in the population. Furthermore, we will construct risk prediction models for early detection of digestive system diseases.
Project Duration: 36 months.
Public Health Impact: Our research findings may establish an effective strategy for early intervention in digestive system diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/combined-effects-of-healthy-lifestyle-factors-for-early-onset-colorectal-cancer-incidence-and-mortality

Combined effects of healthy lifestyle factors for early-onset colorectal cancer incidence and mortality

Last updated:
ID:
63483
Start date:
24 September 2020
Project status:
Closed
Principal investigator:
Dr Hao Bai
Lead institution:
Zhejiang University, China

Summary
Over the past decades, although the overall incidence and mortality of colorectal cancer trends have been declining among the entire population, an increasing trends in the incidence of early-onset colorectal cancer (age!50 years) was recently observed. However, the reason for rising EOCRC incidence and mortality are unclear. EOCRC tumors are different from traditional colorectal cancer in clinical, pathological, and molecular. Currently, a comprehensive analysis of the impact of environmental risk factor on EOCRC based on case-control studies or cohort studies is lacking. In addition, colorectal cancer is a multifactorial disease, requiring a combination of exposures for its development. Single environmental factor is not enough to clarify the interaction between different risk factors. Some risk factors are highly correlated, which is difficult to distinguish their independent effects.
The study purposes are as follows: (1) To estimate the impact of individual lifestyle factors (e.g. body mass index, cigarette smoking, alcohol consumption, physical activity, aspirin use, antibiotics use, and diet factors such as daily intake of fruit/vegetables, daily intake of red and processed meat, daily intake of milk) on early- onset colorectal cancer (EOCRC) incidence and mortality. (2) To assess the combined effects of the healthy lifestyle factors on EOCRC incidence and mortality through using a combined exposure score method based on the number and level of risk factors to which each participant was exposed. (3) To examine whether the impacts of lifestyle factors for EOCRC is different from that of late-onset colorectal cancers.
The estimated duration of this project is 12 months. This study will identify potential risk factors for EOCRC morbidity and mortality, and evaluate the impact of combined healthy lifestyle factors on EOCRC morbidity and mortality. This information could be used by general and high risk population to prevent EOCRC and help to develop prevention and screening strategies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/combined-impact-of-genetic-variants-that-physically-interact-in-the-3d-genome-on-disease-susceptibility

Combined impact of genetic variants that physically interact in the 3D genome on disease susceptibility

Last updated:
ID:
45624
Start date:
20 February 2019
Project status:
Current
Principal investigator:
Dr Olivia Corradin
Lead institution:
Whitehead Institute for Biomedical Research, United States of America

Genetic variation enables the vast diversity observed in the human population, but genetic variation is also what defines our predisposition to disease. One of the primary goals in the field of genetics is to identify which genetic variants contribute to disease in order to learn more about how a disease works and affects the body. Ultimately, understanding these genetic risk factors can help inform patient diagnosis, treatment, and preventative care. However, identifying these genetic variants, is only the first step toward these goals. In order to identify new insights into the pathogenesis of human disease, we must first understand why a particular genetic variant puts one at risk of developing the disease.

There are two critical steps toward understanding why a particular genetic variant contributes to disease. The first is to determine the gene that is affected by the genetic variant and secondly, to determine where in the body this effect impact takes place. These questions are often challenging because many genetic risk factors do not fall within genes. Rather, these genetic variants can be found in “regulatory elements” which do not encode for proteins. These regulatory elements vary across the different cell types in the human body and play an important role in controlling which genes are made into proteins and how much protein is made in any given tissue.

In this study, we leverage new insights about the function of regulatory elements to help determine what tissue is affected by the genetic risk factor. We have previously applied this approach to multiple sclerosis, which is both an autoimmune and neurodegenerative disease. We identified MS genetic risk factors that impact immune cell function in the blood, as well as genetic factors that impact the function of neuron-protecting cells in the brain. Using the UKbiobank, we will apply this approach to the study of neuropsychiatric diseases, in particular substance use disorders. We will integrate results from our analysis of the UKbiobank data with experimental data that measures regulatory element function in different brain cell types and regions. This will enable us to evaluate which cell types in the brain are affected by genetic variation that increases one’s susceptibility to these traits. This study will enable us to generate new information as to why a given genetic variant contributes to disease and will reveal new insights into how a given disease progresses within the body.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/combined-latitudinal-and-longitudinal-study-on-the-impact-of-wildfire-smoke-on-lung-health

Combined Latitudinal and Longitudinal Study on the Impact of Wildfire Smoke on Lung Health

Last updated:
ID:
446074
Start date:
28 September 2025
Project status:
Current
Principal investigator:
Dr Christina Thornton
Lead institution:
University of Calgary, Canada

This study aims to investigate the impact of wildfire smoke on lung health across different geographical regions and over time using a multi-omics approach. By recruiting individuals from various regions with varying exposure levels, and following them over multiple time points, the study will assess immediate and long-term effects on lung health. Biological samples, including blood, nasal swabs, saliva, and exhaled breath condensate, will be collected and analyzed using microbiome sequencing, transcriptomics, proteomics, metabolomics, and epigenomics. The data will be integrated using geospatial and time-series analyses, with machine learning employed to identify biomarkers and predict long-term health outcomes. The study will also validate key findings through qPCR, ELISA, and functional studies in vitro and in animal models. The UK Biobank will enhance this research by providing extensive genetic, imaging, and health data, allowing researchers to explore genetic predispositions and long-term health outcomes related to wildfire smoke exposure, thereby informing personalized healthcare and public health policies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/combined-lifestyle-genetic-susceptibility-and-the-risk-of-accelerated-kidney-function-decline

Combined lifestyle, genetic susceptibility and the risk of accelerated kidney function decline.

Last updated:
ID:
134892
Start date:
21 November 2023
Project status:
Current
Principal investigator:
Dr Yue Shen
Lead institution:
Shanghai Jiao Tong University School of Medicine., China

Chronic kidney disease (CKD), characterized by consistent loss of kidney function for more than three months, affects around 10% of the total population. The influence of overall lifestyle behaviors on renal function, CKD related complications, and deterioration of kidney function remains unclear. To clarify the risk factors of loss of kidney function and the association among them is an important issue. In this study, we aim to investigate the associations of lifestyle factors, genetic susceptibility, as well as the synergies of lifestyles and genetics with risks of accelerated kidney function decline, and to explore the potential effects of metabolites on renal function. We plan to use the cohort data in UK Biobank. Rate of renal function decline of CKD patients will be determined according to biochemicals indicators. Socioeconomic characteristics, biochemical laboratory results, comorbidity, medication, genetic information, lifestyle behaviors such as habitual diet, alcohol intake, smoking status, physical activity and sleeping pattern would be acquired and evaluated. Genetic variants will be investigated to identify subgroups of CKD patients to gain insight into the underlying mechanisms that drive different progression trends. The association between lifestyles, potential related genes and stratified factors on the risk of declined renal function will be analyzed. We also propose to uncover some potential biomarkers mediating the interactions between lifestyle factors and rapid decline of renal function. We hope this 3-year project could help determine the appropriate lifestyles for CKD patients, identify risk factors for rapid deterioration of kidney function, and offer suggestions of interventions that are critical in preventing exacerbation of glomerular filtration rate. We hope that incorporating human genetic evidence and lifestyles into our prevention and therapeutic pipeline could contribute to slow down deterioration of kidney function, thus benefiting more patients.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/combining-brain-physiology-polygenic-scores-and-dietary-intake-patterns-to-better-understand-the-causes-and-consequences-of-obesity-related-diseases

Combining brain physiology, polygenic scores, and dietary intake patterns to better understand the causes and consequences of obesity-related diseases.

Last updated:
ID:
41060
Start date:
6 December 2019
Project status:
Closed
Principal investigator:
Professor Philipp D Koellinger
Lead institution:
VU University Amsterdam, Netherlands

We aim to break new grounds in the understanding of the relationship between eating behavior and brain physiology. Our study will combine the available brain images in the UKB with information about dietary intake, related health-outcomes, and genome-wide data.
The insights from our research will advance the understanding of the biological mechanisms underlying dietary intake, and may identify targets of novel treatments and interventions for eating disorders and obesity. Importantly, our research design will combine neurophysiological measures with behavioral and genetic data that will allow addressing questions regarding the direction of causal relationships between brain physiology, eating behavior, and health.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/combining-deep-neural-networks-and-large-scale-brain-data-to-predict-human-cognition-and-behavior

Combining Deep Neural Networks and Large-Scale Brain Data to Predict Human Cognition and Behavior

Last updated:
ID:
69566
Start date:
22 April 2021
Project status:
Closed
Principal investigator:
Dr Brian Odegaard
Lead institution:
University of Florida, United States of America

In the last decade, Deep Neural Network models (DNNs) have facilitated remarkable advances in brain decoding, revealing how distinct patterns of neural activity correspond to different thoughts, sensations, and behaviors. From brain activity alone, these models can predict what images a person is seeing or imagining in each moment, current states of pain, specific auditory sensations, and many other mental states. When trained on sufficiently large data, they can not only be used to decode current mental states, but also predict specific phenotypes (e.g., average weekly alcohol intake, fluid intelligence, etc.). In this investigation, we will combine DNNs with a “big data” approach to decode cognitive and behavioral phenotypes from existing brain data samples collected at the University of Florida. Specifically, by leveraging the computing power of the HiPerGator supercomputer, we will build predictive models from the large, openly accessible UKBioBank dataset, and then apply these models to predict cognitive and behavioral phenotypes from specific populations of interest (e.g., aging individuals). This work will (i) facilitate development of (and make publicly available) a new research algorithm for deep learning, and (ii) provide insights into how brain structure and function are linked to different phenotypes. The duration of this project is from January 1, 2021, through January 1, 2022. The public health impact of this project is that by using deep learning, we will be able to (i) identify predictive markers for maladaptive phenotypes (e.g., alcohol abuse) that can be used to inform behavioral and therapeutic interventions, and (ii) identify brain regions of interest for future targeted neurofeedback interventions with fMRI, such as using neurofeedback to decrease cigarette, food, and alcohol cravings.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/combining-genetic-analysis-of-disease-related-phenotypes-with-in-vitro-disease-models-for-development-of-high-efficacy-and-low-toxicity-therapeutics

Combining genetic analysis of disease related phenotypes with in vitro disease models for development of high efficacy and low toxicity therapeutics

Last updated:
ID:
51766
Start date:
3 July 2019
Project status:
Current
Principal investigator:
Dr Colm O'Dushlaine
Lead institution:
Insitro, Inc., United States of America

The development of new therapeutics for human diseases is a challenging process with high costs and failure rates. A major cause of these failures is the lack of good pre-clinical in vitro models for many diseases. Recent studies show that investigating the genetic and environmental influences on diseases can guide the drug discovery process by identifying key driver genes, biological processes and cell types. Therefore, we will use the UK Biobank resource to implement and develop new statistical and machine learning approaches for such analysis, with the aim of guiding in vitro disease model development, and identifying and validating new therapeutic targets. We aim to first apply this approach to develop therapeutics for neurological, metabolic and immunological disorders. Considering that our project involves the generation of hypotheses as well as experimental validation of new therapeutics, we expect it to take several years. Nevertheless, a successful outcome of this project can have a significant effect on our ability to develop new, effective therapeutics, and hence on the well-being of many patients.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/combining-genetic-and-environmental-information-to-predict-disease-risk

Combining Genetic and Environmental Information to Predict Disease Risk

Last updated:
ID:
58024
Start date:
2 March 2020
Project status:
Closed
Principal investigator:
Dr Bjarni Vilhjalmsson
Lead institution:
Aarhus University, Denmark

The aim of this project is to develop a new computational approach to predict disease risk of an individual based on various information, including the genome of the individual, family history, medical history, and other information. Such predictions are useful in several different scenarios, both in clinical and research applications. In clinical settings risk predictions are routinely used to identify at-risk individuals, for example, BMI and blood measurements are currently used to assess risk for developing heart diseases. Similarly, genetic tests are commonly used to estimate risk for developing breast cancer. In this context, we believe our work can help improve the accuracy of these risk assessments. However, for us the main motivation for improving risk prediction stems from research applications, and not clinical application. In research settings, understanding risk for developing a specific disease can provide insights into what actually causes the disease, and hopefully, how we can prevent it. Similarly, the predicted disease risk can be used to study the relationship with other diseases and their underlying biology.

The UK biobank dataset is uniquely suited for carrying this work out, as it is an exceptionally rich data resource. It will allow us to consider a wide range of parameters when optimising our predictor, including the genome, health records, and other information when predicting disease risk for an individual. Moreover, using the genome of an individual we can identify other relatives in the UK biobank data, and consider their information as well when predicting the disease risk. The family information can tell us something about the environment that the individual in question lives in.

In terms of specific applications, we will apply the method to study mental health. By predicting risk for developing psychiatric disorders and related medical conditions, we hope our research will eventually provide valuable insights into the underlying causes of these debilitating conditions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/combining-genetic-and-phenotypic-data-in-type-2-diabetes-to-deduce-a-genetic-risk-score

Combining genetic and phenotypic data in type 2 diabetes to deduce a genetic risk score

Last updated:
ID:
42406
Start date:
28 January 2019
Project status:
Closed
Principal investigator:
Dr Ramesh Menon
Lead institution:
MedGenome Labs Pvt Ltd., India

Genome-wide association studies are useful in identifying genetic variants associated with human complex diseases. Polygenic risk score summarizes weights of multiple genetic loci associated with the disease. The goal of PRS is to stratify patients into risk categories based on their genetic mutations. Here, by utilizing the UKBiobank data, we aim to understand this relationship and to derive a polygenic risk score in diabetes, and the relationships between various traits/clinical observations and their associated risks. This new knowledge will lead to the development of new diagnosis, prevention and hopefully the identification of potential therapeutic targets, and improve the health of an aging population.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/combining-genetic-environmental-and-clinical-information-to-predict-the-risk-of-gastrointestinal-cancer

Combining genetic, environmental and clinical information to predict the risk of gastrointestinal cancer

Last updated:
ID:
91799
Start date:
9 August 2022
Project status:
Current
Principal investigator:
Professor Yulian Wu
Lead institution:
Zhejiang University, China

1. Aims: We aim to construct gastrointestinal cancer risk models with both genetic and non-genetic information.

2. Scientific rationale: Gastrointestinal (GI) cancer is a major cause of mortality and accounts for 3.6 million deaths globally every year. The patients often have a poor prognosis due to the lack of effective strategies directed at early detection. Therefore, improvement of survival is heavily reliant on the development of innovative early detection. However, this population lacks recommendations for early detection and screening. Gastrointestinal cancer is a multifactorial disease, and both environmental and genetic factors play a role in its etiology. Previous risk prediction models often apply established environmental and behavioral risk factors to estimate individual risk. Recently, several studies have included genetic risk factors and non-genetic factors to assess cancer risk more accurately. However, the value of combining genetic and non-genetic factors to predict the risk of gastrointestinal cancer has not been fully explored.

3. Project duration: Three years will be needed to complete this project.

4. Public health: The project is expected to quantify the importance of genetic and non-genetic factors in the individual cancer risk prediction. The findings will improve our understanding of cancer risk assessment model construction and therefore guide us to identify high-risk individuals more accurately. This research will also make important contribution to the guidance of personalized prevention of gastrointestinal cancer by Identifying people who would benefit from secondary screening.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/combining-genetic-information-with-population-imaging-to-improve-diagnostic-markers-for-alzheimers-disease

Combining genetic information with population imaging to improve diagnostic markers for Alzheimer’s Disease

Last updated:
ID:
65299
Start date:
11 January 2021
Project status:
Current
Principal investigator:
Dr Andre Altmann
Lead institution:
University College London, Great Britain

As we age several properties of our bodies change over time as well. For instance, muscles loose strength and brains shrink. Brain disorders, such as Alzheimer’s Disease, lead to brain shrinkage that is much faster than what we would expect in people without the disorder. A disease often does not affect the whole brain the same way. For instance, in Alzheimer’s Disease a small brain region, the hippocampus, shows shrinkage early in the disease. The hippocampus is part of the brain that is particularly important for memory, thus leading to the well-known memory problems in Alzheimer’s disease.

Neuroimaging enables us to take pictures of the brain and measure its size and also the size of the hippocampus. Now, in order to understand whether a person’s measurement is in the healthy range, we can compare their measurement to the ones obtained from age-matched healthy individuals. If we collect such measurements for different age groups we can generate a diagram that tells us how this measure changes in the population over time: a nomogram.

Nomograms are a widely used tool in medicine. Most prominently they are employed to assess and track the growth of newborns. Likewise, a nomogram for hippocampal volume can be used to assess and track the decline of this important part of the brain. People whose measurements are ‘below the curve’ may require further check-ups and closer monitoring. Traditionally, nomograms are generated for men and women separately. However, this is only one possible difference. One other component is genetics. Our genes can influence whether we have naturally a larger or smaller hippocampus. But so far, the contribution of genetics has not been leveraged in the development of nomograms. As a consequence, people with a smaller hippocampus may incorrectly be diagnosed with possible Alzheimer’s disease, whereas people with a naturally larger hippocampus may not be recognized as abnormal until very late in the disease.

In this project we aim to improve nomograms by incorporating genetic information. We will test whether these improved nomograms can better detect abnormal aging and may lead to the earlier diagnosis of Alzheimer’s disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/combining-genetic-profiles-multi-omic-data-and-empirical-observations-via-epidemiology-is-crucial-for-deepening-our-understanding-of-cancers-pathogenesis

Combining genetic profiles, multi-omic data, and empirical observations via epidemiology is crucial for deepening our understanding of cancer’s pathogenesis.

Last updated:
ID:
570157
Start date:
18 February 2025
Project status:
Current
Principal investigator:
Dr De-Huan Xie
Lead institution:
Guangdong Provincial People's Hospital, China

Research Questions:
1. What novel risk factors contribute to various types of cancer?
2. Can we identify new biomarkers for early detection and treatment response?
3. How do genetic, metabolomic, proteomic, and epidemiological factors interplay in cancer development?

Objectives:
– Unearth risk determinants for solid tumors and hematologic cancers.
– Identify prospective biomarkers for early cancer detection and prognosis.
– Elucidate etiological links among genetic, metabolomic, proteomic, and epidemiological data in cancer pathogenesis.

Scientific Rationale:
Cancer is a leading global health threat, with GLOBOCAN 2022 estimating 1 in 5 people will develop cancer. Our understanding of cancer is limited, necessitating a comprehensive approach. This project will leverage UK BioBank’s rich dataset, integrating genetic, metabolomic, proteomic, MRI, and epidemiological data. By applying a multifaceted analytical approach, we aim to deepen our understanding of cancer complexity, tailor interventions, refine diagnostics, and bolster management protocols, ultimately improving cancer-related health outcomes.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/combining-imaging-data-with-genomic-and-phenotypic-data-to-improve-the-whole-course-management-of-human-noncommunicable-diseases

Combining imaging data with genomic and phenotypic data to improve the whole-course management of human noncommunicable diseases

Last updated:
ID:
151193
Start date:
28 February 2024
Project status:
Current
Principal investigator:
Professor Shenghong Ju
Lead institution:
Southeast University, China

Noncommunicable diseases (NCDs), mainly including cardiovascular disease, tumours, chronic respiratory disease and diabetes, are the biggest health problem worldwide. They result from a combination of genetic, physiological, environmental and behavioural factors. Thus, precise identification of risk factors and a deep understanding of pathogenesis are necessary in the prevention, detection, and treatment of NCDs. Current tools that aim to improve the management of NCDs are based on simple measurements and markers and do not work well. In the following three years, we intend to identify the risk factors of the development and the progression of NCDs, explore the imaging value in the interpretation, detection and treatment of NCDs, and combine multi-dimensional variables (including genetic, phenotypic, and imaging data) to promote the diagnosis and treatment of NCDs. As radiologists, we will also sufficiently investigate the essential correlations between imaging data and with occurrence and progression of NCDs, especially based on the genetics and corresponding pathophysiological mechanisms. Our work will provide new insights into public health research for disease prevention, diagnosis and treatment of NCDs, and should assist medical practitioners and society in general by improving healthcare decision-making.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/combining-machine-learning-and-statistics-to-understand-the-genetic-architecture-of-mri-derived-anatomical-structures-and-develop-disease-risk-models-using-these-associations

Combining machine learning and statistics to understand the genetic architecture of MRI-derived anatomical structures and develop disease risk models using these associations.

Last updated:
ID:
87255
Start date:
7 November 2022
Project status:
Current
Principal investigator:
Dr Catherine Tcheandjieu
Lead institution:
J. David Gladstone Institutes, United States of America

We aim to combine machine learning and genomics to understand how changes in our genome impact the anatomy of the cardiovascular system (heart and blood vessels), and how this can lead to cardiovascular diseases (CVD). The diagnosis of CVD relies on the identification of clinical symptoms that can take years to manifest, by which time the disease is often severe and fatal. These symptoms often reflect anatomical damage accrued over time in organs and tissues throughout the body. Some of this anatomical damage can be diagnosed by body imaging such as Magnetic Resonance Imaging (MRI). While MRI diagnosis is better than waiting years for CVD symptoms to appear, it is still not soon enough to capture the disease at the very early stage. We hypothesize that gene variants that contribute to the anatomy of the cardiovascular system can be leveraged to predict risk for CVD. Here, we will use machine learning to extract anatomical features of the heart and blood vessel from cardiac, abdominal, and brain MRIs from healthy individuals and individuals with CVD. We will then correlate these features to the rich genomic data of the UK biobank. This comparison will reveal the extent to which genetic variation affecting the anatomy of the cardiovascular system can help better understand and predict the risk of CVD such as aneurysms, heart failure, and stroke.

Our second project aims to identify genes that predispose to autism spectrum disorder (ASD) and congenital heart defects (CHD). CHD and ASD are the two most common diseases among children. Their high heritability suggests a strong genetic contribution, yet the underlying genes remain unclear. To tackle this question, we concentrate on regions of the genome where variants that affect the 3D structure of the genome have biological consequences such as changes in the regulation of nearby genes. We have developed genomic tools that can predict the 3D configuration of the genome and can identify regions in the genome that are jointly transmitted from parent to children more often than would be expected by chance alone. We will quantify how these regions contribute to the risk of ASD and CHD. Ultimately, this work will reveal genome variants that could be predictors of disease.

We anticipate that our project will take 3 years to complete. Our findings will significantly advance our understanding of genetic risk for diseases and help develop models for risk prediction and therapies beneficial for patients.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/combining-multiple-or-high-dimensional-biomarkers-to-improve-accuracy-for-detecting-covid-19-virus-infection-and-antibody

Combining multiple or high-dimensional biomarkers to improve accuracy for detecting COVID-19 virus infection and antibody

Last updated:
ID:
86711
Start date:
10 August 2022
Project status:
Current
Principal investigator:
Dr Aiyi Liu
Lead institution:
NI of Child Health and Human Development, United States of America

In recent decades, powerful biomarkers have become an important tool in diagnosis to identify subjects with a disease, or at high risk of developing the disease. However, single biomarker is not accurate enough and may incur considerable false positives and false negatives which will have serious implications on patients and public health. Thus, researchers have considered the problem of combining biomarkers to improve diagnosis of a disease. To date, the coronavirus SARS-CoV-2 that causes the severe acute respiratory syndrome COVID-19 was reported to have infected over 380 million people, leading to over 5 million deaths worldwide. Better and faster COVID-19 test for virus infection or antibody is urgently needed to help guide the improvement of detection and treatment. Therefore, our aim of this project is to find more effective and quicker ways for better diagnosis and prediction of COVID-19 virus infection and antibody, using the existing data available in the UK Biobank database on biomarkers and test results for COVID-19 virus infection and antibody. The diagnostic methods developed from this research project will substantially improve the accuracy for detection of COVID-19 virus and antibody, while maintaining the advantage of reducing cost and time for screening.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/combining-nmr-metabolomics-and-data-analytics-to-explore-the-biochemical-basis-of-schizophrenia-pathophysiology

Combining NMR metabolomics and data-analytics to explore the biochemical basis of schizophrenia pathophysiology

Last updated:
ID:
104659
Start date:
19 July 2023
Project status:
Current
Principal investigator:
Dr Abhishek Cukkemane
Lead institution:
Research Center Juelich, Germany

Schizophrenia is a debilitating psychotic disorder that affects 1% of the global population. Currently, no laboratory tests are available to specifically diagnose schizophrenia. Psychiatrists perform the diagnosis and later prognosis of patients based on symptoms as outlined in international medical guidelines such as ICD-10, which is proposed by the World Health Organization.
The biggest bottle neck in such a complex disease are the unknown factors and the interplay of them, which include mental, physical, genetic and environmental conditions. Therefore, many a scientist, including us, apply functional genomics approaches such as metabolomics and proteomics to understand the molecular mechanisms of the disorders. The strength of functional genomics lies in the fact that the technology can be applied to describe how the individual components of a biological system work together to define an individual. In this manner, when one can analyze data from subjects and compare it with healthy controls from a population of people. Based on the differences between the two groups, one is in a better position to identify relevant biological cues for the disorder. For this purpose, we are approaching the UKbiobank requesting access to their data so that we can annotate and quantify for biochemicals that are present in the blood (plasma) samples. This will be followed by performing statistical analysis to identify key reported molecules and the biochemical pathways that are malfunctioning in the disorder.
In the next step, we will extend our approach from identifying the biomolecules to applying the findings to develop diagnostic kits. The need of the hour is a reliable molecular diagnostic tool that can aid the psychiatrists with rapid diagnosis of the diseases. tics along with machine learning approaches to characterize the heterogeneous combination of symptoms observed in schizophrenia over the traditional diagnostic approaches that have proven to be less-effective. To meet our objectives, we perceive a minimum of 36 months and up to 60 months for successful implementation of the project.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/common-and-rare-genetic-variants-associated-with-cardiometabolic-traits-and-disease

Common and rare genetic variants associated with cardiometabolic traits and disease

Last updated:
ID:
70653
Start date:
14 February 2022
Project status:
Current
Principal investigator:
Dr Daniel J Rader
Lead institution:
University of Pennsylvania, United States of America

Cardiometabolic diseases such as cardiovascular disease, metabolic liver disease, diabetes mellitus and hypertension have a complex genetic etiology and often co-occur, which makes distinguishing causal from correlated risk factors very difficult. We, therefore, aim to improve cardiometabolic health by utilizing several modern epidemiological concepts. The premise of this proposal is to understand the effects of common and less common genetic variants on cardiometabolic traits and diseases. The objectives of the proposed project are to identify causal risk factors for cardiometabolic traits, diseases, response to therapy, and outcomes and the effect of genetic variants on those. We will investigate the interaction between genetics and environmental factors and evaluate whether disease prediction can be improved by incorporating this information into novel risk models for cardiometabolic diseases.
The proposed research will significantly improve knowledge of the biological mechanisms that lead to cardiometabolic disease, refine disease prevention and detect new types of markers for early detection and/or intervention. This information will help healthcare professionals advance treatment and prevention strategies for various cardiometabolic outcomes. We expect the scope of the project we have outlined to take three years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/common-and-rare-genetic-variants-in-respiratory-health-the-uk-biobank-lung-exome-variant-evaluation-uk-bileve-consortium

Common and rare genetic variants in respiratory health: the UK Biobank Lung Exome Variant Evaluation (UK BiLEVE) consortium

Last updated:
ID:
648
Start date:
1 November 2012
Project status:
Current
Principal investigator:
Professor Martin Tobin
Lead institution:
University of Leicester, Great Britain

Lung function is an important indicator of respiratory health and mortality. Measures of lung function show irreversible airway obstruction in chronic obstructive pulmonary disease (COPD), a progressive condition affecting 900,000 people in the UK. Smoking is a strong risk factor for COPD but not all smokers are equally susceptible. Genetic approaches to understanding the mechanisms underlying the maintenance of good lung function in some people, and underlying the development of COPD in others, aim to reveal previously unknown molecular targets for drug development and to facilitate stratified approaches to treatment and care. This project aims to detect rare genetic variants associated with lung function. Once discovered, such variants would be very useful tools for the scientific community, because such variants tend to exert a large effect on disease risk and provide a means to translate findings from genetic studies of lung function to clinical relevant research and development. The proposed study leverages the power of UK Biobank and the resources and experience of an expert group of UK collaborators in respiratory genomics to advance understanding of lung function and COPD. This project will use a customised respiratory exome chip in 50,000 UK Biobank participants, selected according to their smoking history and lung function status at baseline. This project therefore requires the use of data (spirometry, smoking and other lifestyle factors) and DNA samples.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/common-and-rare-genetic-variation-in-health-and-kidney-disease

Common and rare genetic variation in health and kidney disease

Last updated:
ID:
98596
Start date:
8 August 2023
Project status:
Current
Principal investigator:
Professor Daniel Gale
Lead institution:
University College London, Great Britain

The UK Biobank represents a rich data trove that incorporates health and lifestyle information about a large group, thought to be representative of the general UK population. We aim to use this information to answer the question of what drives a variety of diseases affecting the kidneys.
1. We will look for differences between the genes (instructions our body needs to develop and function) of people with and without evidence of different types of kidney disease to find out more about the causes of these conditions, including why some people are affected more severely than others.
2. For kidney diseases with many complex causes, we will look at what proportion of that disease could be attributed to genes. How are these genes causing this disease?
3. We will also take genes which are known to cause rare kidney diseases, and study whether they contribute to a lesser degree towards more common diseases. For example, we will explore the impact of genes associated with kidney disease on outcomes such as heart disease.
We hope the results of this study will enable doctors to make more accurate diagnoses and offer better treatments to people with or at risk of kidney disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/common-and-rare-genetic-variations-in-autoimmune-diseases

Common and rare genetic variations in autoimmune diseases

Last updated:
ID:
197617
Start date:
5 November 2024
Project status:
Current
Principal investigator:
Dr Yiming Luo
Lead institution:
Columbia University, United States of America

Autoimmune diseases are a group of disorders characterized by an abnormal immune response leading to illness in various organs. There is an unmet need to understand the contribution of genetic changes to these disorders. Our long-term goal is to identify the causes of these disorders and develop personalized treatment approaches. Our study aims include: 1. Understanding the contribution of common and rare genetic variants in autoimmune diseases. 2. Prioritizing putative causal genes mediating the connection between genetic variants and autoimmune diseases. 3. Predicting an individual’s risk of developing autoimmune diseases based on their genetic information. The project duration is three years. This study may help to better understand the causes of autoimmune diseases and facilitate the development of targeted treatments in the future.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/common-and-rare-traf3-variants-influence-b-cell-survival-and-immunoglobulin-levels

Common and rare TRAF3 variants influence B cell survival and immunoglobulin levels

Last updated:
ID:
53026
Start date:
6 April 2020
Project status:
Closed
Principal investigator:
Dr William Rae
Lead institution:
University of Cambridge, Great Britain

BACKGROUND: Autoimmune diseases are conditions in which the immune system attacks a patient’s own body. Autoimmune diseases effect approximately 1 in 12 people, and this number is increasing, with no cure in the majority of cases. The reason autoimmune diseases develop is not fully understood with genetic and environmental factors playing roles. Many large studies have identified genetic changes in the gene TRAF3 as increasing the risk of developing autoimmune diseases. We have also recently identified very rare changes in the gene TRAF3 as causing autoimmunity in some individuals. In the general population there are many common genetic changes seen in the gene TRAF3 and we will investigate if the presence of any common changes alter the risk of developing an autoimmune disease.

AIM: To assess the impact of common changes in TRAF3 on autoimmunity risk.

SCIENTIFIC RATIONALE: The gene TRAF3 controls several functions of the immune system. Some people within the general population have changes in this gene which may effect how well it works, and in doing so alter the risk of developing autoimmune disease. We plan to study how changes in the TRAF3 may increase the risk of developing autoimmune disease. This is important information as specific treatments which target the functions of this gene have been developed and may provide new treatments for patients with autoimmune diseases and genetic changes in the gene TRAF3.

PROJECT DURATION: The data analysis and testing of these TRAF3 changes are expected to take 24 months.

PUBLIC HEALTH IMPACT: Autoimmune diseases are life-long incurable conditions, and are increasing in the population. Autoimmune disease is a significant economic burden on the health system with treatments needing continued monitoring due to side effects or lack of effect. Understanding how TRAF3 genetic changes may increase the risk and drive autoimmune disease also has the potential to lead to new treatments of autoimmune disease tailored to the genetic make-up of an individual.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/common-genetic-and-neuroimaging-mechanisms-between-insomnia-and-substance-use-disorder

Common genetic and neuroimaging mechanisms between insomnia and substance use disorder

Last updated:
ID:
69196
Start date:
30 March 2021
Project status:
Current
Principal investigator:
Professor Jie Shi
Lead institution:
Peking University, China

Both insomnia and substance use disorder cause significant public health and socioeconomic burden. People with substance use disorder are likely to comorbid with insomnia, and individuals who have sleeping problems may abuse alcohol or illicit drugs or prescription medications. The mechanisms underlying altered sleep performance following the substance use disorder process are unknown. The neural basis of the increased functional connectivity for the substance use disorder with insomnia is unknown neither. We could identify neural and genetic mechanisms that mediate the association of insomnia and substance use disorder. The whole program will last about 3 years. It will help us to develop new strategies of treatment for insomnia symptoms in patients with substance use disorder.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/common-mental-disorder-and-its-correlates-in-uk-biobank

Common Mental Disorder and its Correlates in UK Biobank

Last updated:
ID:
34553
Start date:
25 June 2018
Project status:
Current
Principal investigator:
Professor Matthew Hotopf
Lead institution:
King's College London, Great Britain

Using the mental health questionnaire, baseline data, and data linkage we will characterise mental disorder in the whole UKB with a view to identifying phenotypes of common mental disorder (CMD), taking into account symptoms, longitudinal course and severity. We would like to use actigraphy data to further characterise phenotypes of CMD in this subset. CMD often co-occur with physical disorder, sensory and cognitive impairment. We would like to study the association with CMD as risk factor and outcome. We will examine suggested biological correlates of CMD in biomarkers and genotypes. Over one third of participants in the UK Biobank (UKB) are likely to have suffered from a common mental disorder (CMD), at some point in their lives, including depression, anxiety and alcohol misuse. We will estimate the impact of CMD on health and well-being, and explore targets for further study. People with severe mental illness die earlier than their peers due to physical disease, associated with less treatment for their physical disease. We would like to see how this extends to CMD. Insights may lead to new opportunities improving health. We will need access to information from baseline (history/examination), the mental health questionnaire and data linkages. We will use conventional mental health diagnostic categories, and also use cluster analysis to investigate other ways of understanding mental disorder. We will perform regression analysis (tests of association) between physical and mental health categories, taking into account all psychosocial data and health behaviours (eg diet and exercise) including potentially sensitive areas of sexuality (baseline) and self harm (mental health questionnaire). Full


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/common-risk-factors-and-mechanisms-in-cardiometabolic-diseases-and-neurodegenerative-diseases

Common risk factors and mechanisms in cardiometabolic diseases and neurodegenerative diseases

Last updated:
ID:
55005
Start date:
14 January 2020
Project status:
Current
Principal investigator:
Professor Geng Zong
Lead institution:
Shanghai Institute of Nutrition and Health, China

Aims: Our study aims to uncover potential links between cardiometabolic diseases and neurodegenerative diseases, by focusing on their shared risk factors and mechanisms. Scientific rationale: These two categories of diseases affect a large and growing population worldwide, and current studies suggest that they are interconnected. For example, unfavorable lifestyles (such as smoking and alcohol drinking) and mutations in key genes (such as APOE genes) may increase risks of both cardiometabolic diseases and neurodegenerative diseases. Meanwhile, hyperglycemia, hypertension, and dyslipidemia have been associated with structural brain abnormalities and higher dementia risk in later life, whereas Alzheimer’s disease-related proteins has also been suggested to promote diabetes. A systematic study on their shared risk factors and mechanisms will deepen our understanding on their pathogenesis and progression. Project duration: This complicated project includes several independent analyses and requires close collaborations between two research groups. We expect to achieve some initial milestones in the next 3 years. Public health impact: Our study may promote public health policy priorities by focusing on common environment risk factors of two major disease categories. Evidences on the bidirectional relationship and common pathways of these diseases may inform healthcare providers to ensure the long-term survival of patients with either health conditions, and inspires new clinical practices in disease treatment.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/common-risk-factors-and-prognosis-of-inflammatory-bowel-disease-and-irritable-bowel-syndrome

Common risk factors and prognosis of inflammatory bowel disease and irritable bowel syndrome

Last updated:
ID:
74444
Start date:
20 July 2021
Project status:
Current
Principal investigator:
Professor Shanshan Wu
Lead institution:
Beijing Friendship Hospital, Capital Medical University, China

Aims: Our study aims to investigate potential risk factors and prognosis of inflammatory bowel disease and irritable bowel syndrome diseases. Scientific rationale: These two categories of diseases affect a large and growing population worldwide. However, the potential lifestyle and environment risk factors are not exactly clear until now. Current studies suggest that some lifestyle factors, such as diet and physical activity, may play an important role in the development of these two categories of diseases. Meanwhile, the prognosis of these two categories of diseases, especially the long-term prognosis is still lack of evidence. Whether inflammatory bowel disease and irritable bowel syndrome diseases are associated with increased risk of multiple health-related outcomes, such as cancer, metabolic diseases, mental disorders and mortality, is still needed to investigate and assess in the large-scale, prospective, long-term cohort study. Project duration: This complicated project includes several independent analyses. We expect to achieve some initial milestones in the next 3 years. Public health impact: Our study may promote public health policy priorities by focusing on common lifestyle and environment risk factors of inflammatory bowel disease and irritable bowel syndrome diseases. Evidences on the risk factors and prognosis of these diseases may inform healthcare providers to ensure the long-term survival of those patients, and inspires new clinical practices in disease treatment.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comorbidities-among-mental-and-physical-disorders-using-data-from-genome-wide-association-studies-gwas

Comorbidities among mental and physical disorders using data from Genome-Wide Association Studies (GWAS)

Last updated:
ID:
58200
Start date:
26 August 2022
Project status:
Closed
Principal investigator:
Professor Ian Gizer
Lead institution:
University of Missouri, United States of America

Individuals diagnosed with substance use and addiction-related disorders often meet diagnostic criteria for one or more additional mental health and/or physical health disorders. Taken together these health concerns result in substantial economic burden in the form of public health costs each year. Research aimed at understanding what causes and influences these disorders has become increasingly important and productive.

Specifically, recent advances in the field of genetics and the growing feasibility of large-scale research collaborations has allowed rapid improvements in the identification of genetic influences associated with mental and physical health disorders. The last decade has seen vast improvements in the time and effort required to generate comprehensive genetic data for individuals and this has led to large genetic datasets through initiatives like the UK Biobank and private companies such as 23andMe.

The aim of the current study is to use these large datasets and advanced statistical genetics approaches to investigate the extent to which measured genetic variation contributes, individually or together with other related traits, to the development of addiction and related mental (e.g., antisocial personality disorder) and physical health (e.g., chronic pain, obesity) conditions. The methods of this study will also allow researchers to test how specific genes or sets of genes may be causally related to specific addiction outcomes and related physical or mental health problems (e.g., problematic drinking resulting from chronic pain). The long-term aim of the proposed study is to aid in the development of precision medicine by characterizing the biological mechanisms that increase risk for these disorders in order to advance tailored prevention and treatment efforts. More immediate public health benefits of such research include the value of contributing to a knowledge base that can help guide future data collection and analysis focused on the development of genetic assessments related to risk for these disorders.

The proposed duration for this project is 36 months. This duration will allow adequate time to obtain and analyze genetic datasets and publish results from these analyses in peer-reviewed journals. The proposed methods and analyses of the current project involve using state-of-the-art statistical genetics approaches to estimate the shared genetic influences of these disorders at the level of single genetic variants, individual genes, and sets of genes representing biological pathways related to addiction.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comorbidities-and-non-allergic-outcomes-in-atopic-dermatitis

Comorbidities and Non-allergic Outcomes in Atopic Dermatitis

Last updated:
ID:
122349
Start date:
5 October 2023
Project status:
Current
Principal investigator:
Professor Jianjun Qiao
Lead institution:
Zhejiang University, China

Atopic dermatitis (AD) is a chronic inflammatory disease that can cause pruritic, polymorphic skin lesions. As it is a relapsing disease, treatment aims to alleviate clinical symptoms, eliminate predisposing or exacerbating factors, and reduce flares and comorbidities. Several recent studies have indicated that patients with AD may have a higher risk of developing chronic diseases such as cardiovascular disease compared to non-AD patients. In order to systematically investigate comorbidities and long-term outcomes in AD patients, we will complete this project within 36 months after all the data have been collected. Our project will involve interdisciplinary research teams with epidemiological, clinical, and global health expertise. It is hoped that our findings would help improve the management of AD patients.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comorbidities-and-systemic-inflammation-related-outcomes-of-inflammatory-bowel-disease

Comorbidities and Systemic Inflammation Related Outcomes of Inflammatory Bowel Disease

Last updated:
ID:
73595
Start date:
5 July 2021
Project status:
Closed
Principal investigator:
Dr Jie Chen
Lead institution:
Zhejiang University, China

Inflammatory Bowel Disease (IBD) is a group of disorders that causes sections of the gastrointestinal tract to become severely inflamed and ulcerated. In the absence of a cure, the lifelong conditions and the goal of therapy is to reduce flares of disease and the complications from long-term inflammation. Some recent studies pointed out the higher risk of some chronic diseases in IBD patients like cardiovascular disease and dementia compared with non-IBD people. To systematically investigate the comorbidities and long-term outcomes of IBD patients, we will complete this project within 36 months after all data have been obtained. This proposed project forms a collaborative project led by a multidisciplinary team of researchers with expertise in epidemiology, clinical medicine, and global health. With this project, we aim to provide evidence useful for developing better management of inflammatory bowel disease patients.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comorbidities-between-addiction-and-mental-disorders-association-of-genetics-biomarkers-and-environment-e-g-lifestyle-with-mental-health-and-cognitive-function

Comorbidities between addiction and mental disorders ? association of genetics, biomarkers and environment (e.g. lifestyle) with mental health and cognitive function

Last updated:
ID:
30172
Start date:
6 April 2018
Project status:
Current
Principal investigator:
Professor Helgi Schiöth
Lead institution:
Uppsala University, Sweden

We aim to investigate interactions between genetics and environmental factors e.g. lifestyle in relation to mental health and addiction. We will examine whether environmental and other risk factors (psychiatric comorbidities) influence addiction besides having overlapping genetic risk factors crucial for prevention and treatment strategies, study genetic variants with known associations to addictions and psychiatric disorders and perform genome-wide exploratory analyses for novel variants. We will investigate the association and modulating effects of genetic variation between mental health and addiction, including pharmacogenetics, known and novel biomarkers, SNPs role in drug effects and cognitive function to understand susceptibility to addiction. Addictions and drug abuse are severe common psychiatric disorders, among the leading causes of morbidity and preventable mortality, and often linked with other psychiatric diseases as consequences or potential cause of addictive behavior (Agrawal, 2012). We will evaluate the impact of genetic and environmental risk factors on comorbidities of addiction and common psychiatric disorders. Better understanding how neural circuitry, genetics and environmental factors interact in the etiology of addiction and other psychiatric diseases offer better treatment strategies. Novel insights into molecular biology, genetic, pharmacogenetics and epigenetic mechanisms underlying these associations will improve public-health related decisions for health professionals.
Statistical models will be utilized to analyze associations between genotype, cognitive function and mental health while controlling for demographics such as age, gender, medications, personality traits, physical activity, etc. as well as comorbidities. To examine causal effects of these genes, Mendelian randomization will be applied. Novel genetic variants will be derived from two-thirds of the cohort and validated in the remaining third. Linear models will be used to study the association between environmental factors, cognitive function and addictive behavior. Modifying effects of biomarkers on cognitive function and mental health will be assessed by incorporating it through generalized linear models. To maximize power, the full cohort will be included in the project. We would also like to include the new data from additional participants (still to be released).


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comorbidity-and-covariation-of-indicators-for-disorders-impacting-behaviour-and-mental-health

Comorbidity and covariation of indicators for disorders impacting behaviour and mental health

Last updated:
ID:
101294
Start date:
5 December 2023
Project status:
Current
Principal investigator:
Dr Michel Belyk
Lead institution:
Edge Hill University, Great Britain

Public mental health is a growing cause for concern. In public discourse “Mental Health” often refers to some combination of chronic anxiety or depression. However, biomedical research makes finer distinctions into separate disorders and conditions. Being specific in this way helps researchers to uncover the causes of specific conditions, and helps clinicians to develop appropriate treatments.

However, it is becoming increasingly clear that many of these conditions are comorbid – that is, they tend to occur together. For example, a person who is diagnosed with an anxiety disorder is also more likely to be diagnosed with major depression. Similar patterns of comorbidity are observed with many common disorders of mental health or behaviour including autism spectrum disorder, attention deficit hyperactivity disorder, dyslexia, schizophrenia, substance abuse, eating disorders, and developmental communication disorders, among others.

These high rates of comorbidity are a source of difficulty for researchers trying to uncover the mechanisms of these disorders. Considering patients with multiple diagnoses can lead the causes of one disorder to be confused with another, while excluding patients with multiple diagnoses ignores an important component of the mental health landscape and one where research attention is acutely needed. Moreover, either of these approaches make it difficult to discover why these conditions are comorbid in the first place.

This project proposes to replicate previous findings about the causes of mental health and behavioural disorders about which members of the research team have individual expertise, and assess the degree to which patterns of comorbidity can be attributed to shared genetic, neurological, or environmental factors.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comorbidity-and-lifestyle-on-the-outcome-of-cardiovascular-diseases

Comorbidity and lifestyle on the outcome of cardiovascular diseases

Last updated:
ID:
280417
Start date:
25 October 2024
Project status:
Current
Principal investigator:
Professor Ronghui Yu
Lead institution:
Anhui Medical University, China

Aim:
To explore the effects of genetic and environmental factors on cardiovascular diseases.
Scientific rationale:
Cardiovascular diseases (CVD) are one of the leading causes of death worldwide. However, our understanding of the pathophysiology of CVD is limited. The advances in omics may facilitate a deeper understanding of the process and interaction involved in CVD.
Project duration:
Three years
Public health impact:
This project aims to provide more insights into the development and progression of CVD, which is crucial to establish more effective early intervention strategies and to reduce the CVD burden.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comorbidity-of-major-noncommunicable-diseases-and-mental-disorders

Comorbidity of major noncommunicable diseases and mental disorders

Last updated:
ID:
65418
Start date:
17 May 2021
Project status:
Current
Principal investigator:
Dr Yawei Guo
Lead institution:
Sun Yat-Sen University, China

Major noncommunicable diseases (NCDs) generally include cardiovascular diseases (e.g. coronary heart disease and stroke), diabetes mellitus, cancers and chronic obstructive pulmonary diseases. They are the leading causes of disease burden and mortality globally. Previous research has shown that NCDs and mental disorders are closely related with each other. First, epidemiological studies have consistently showed the commodity of NCDs and mental disorders. Second, patients with NCDs are at higher risk of having and developing mental disorders, and vice versa. Third, both NCDs and mental disorders are associated with increased risk of mortality. Although the associations major NCDs and mental disorders has been well-recognized, the interactions between the two are usually overlooked in clinical practice. In addition, the comorbidity should be further investigated with a large sample size, in order to find the key modifiable risk factors that could be used in prevention and intervention program.

With this 3-year project using UK Biobank data, we will explore the risk factors of the comorbidity of NCDs and mental disorders. The findings of our project will contribute to a better understanding of the comorbidity of NCDs and mental disorders, and promote a more integrated disease management strategy for patients with comorbidity. It also provides evidence on the modifiable risk factors that should be focused in interventions in order to reduce the risk of developing comorbidity in adults.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comorbidity-study-of-brain-health-related-diseases

Comorbidity study of brain health-related diseases

Last updated:
ID:
200220
Start date:
7 August 2025
Project status:
Current
Principal investigator:
Dr Yi Guo
Lead institution:
Second Affiliated Hospital of Zhejiang University, China

The human brain is the command centre for the nervous system. Maintaining a healthy brain during one’s life is the uppermost goal in pursuing health and longevity. As the population ages, the burden of neurological disorders and challenges for the preservation of brain health increase.
We pay special attention to the comorbidity of different brain health-related diseases (such as epilepsy, cerebrovascular diseases, Alzheimer’s disease, and mental disorders, etc.), and the comorbidity of brain health-related diseases with other physical disorders (such as cardiovascular disease, respiratory diseases, digestive diseases, obesity and endocrine diseases, rheumatic diseases, tumors, etc.).
We plan to use the large-sample, dynamic follow-up cohort data of UK Biobank to explore common clustering patterns and influencing factors of multiple comorbidities; summarize the dynamic changes in the psychological, physiological and social spiritual needs of patients at different stages of chronic diseases to achieve precise prevention and management.
The proposed program will last for three years but it might be prolonged due to advances in methodology and novel findings which may require external validations.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comparative-analyses-of-clinical-phenotypes-and-genetic-predisposition-for-cancer-and-emotional-vulnerability-across-different-ethnic-populations

Comparative analyses of clinical phenotypes and genetic predisposition for cancer and emotional vulnerability across different ethnic populations

Last updated:
ID:
54423
Start date:
11 March 2020
Project status:
Closed
Principal investigator:
Professor Eric Y Chuang
Lead institution:
National Taiwan University, Taiwan, Province of China

Mood disorders are potential risk factors that reduce survival for cancer patients. Amongst others, anxiety, neuro-cognitive-dysfunction, sexual-dysfunctions, sleep-disturbance, stress-related disorders/PTSD, suicidal-tendencies, and bipolar and obsessive-compulsive disorders are symptoms that has been observed in cancer patients/survivors. Also, people with severe mental illness, including depression, are less likely to receive routine cancer screening. Therefore, there has been less work on the effect of psychiatric illness on cancer prognosis and survival. It has been found that a subset of cancer patients continue to be vulnerable to this complication even after treatment has ended, and often have difficulties with multitasking, short-term memory, word-finding, or attention. The underlying mechanism is not fully elucidated but may include direct neurotoxic effects of therapy, oxidative damage, and genetic predisposition. Lifestyle and environment of the patients are also important parameters that need to be investigated as a potential risk factor. Therefore, we propose to utilize data from different populations in a period of 3 years to examine the associations between mental health problems and cancer risk factors. The first year will be utilized to examine the factors that are potentially associated with risk of developing cancer for patients with mental health issues and to check for stratification effect due to sex, age and race for specific cancer types. Also demographic characteristics, health status, lifestyle effects and cancer risk factors will be compared for different mental health subgroups. Furthermore, genomic data will be used to conduct association tests to detect significantly associated SNP/CNVs and for genetic correlations amongst different cancer and mood-related conditions. The second year will be used to conduct survival analysis for patients with respect to different demographic, clinical and genomic factors. Also significantly associated findings from first year and imaging data would be used to train deep-learning models to predict survival. Finally, in the third year, comparison studies will be done between UK biobank and Taiwanese samples to establish ethnicity specific findings. As psychiatric patients have less likely, been receiving, specialist procedures, than general population, not adequate data is available for such studies. Therefore, it’s difficult to predict mental health impact on cancer diagnosis and treatment. This study will contribute on establishing factors that has an effect on the emotional vulnerability and comorbidity of mental health and cancer, among patients, thereby providing necessary information that has been lacking in this field of study.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comparative-analysis-of-current-health-risk-prediction-capabilities

Comparative analysis of current health risk prediction capabilities

Last updated:
ID:
199268
Start date:
30 April 2024
Project status:
Current
Principal investigator:
Dr Péter Mendik
Lead institution:
XUND Solutions GmbH, Austria

Comparative analysis of current health risk prediction capabilities
As non-communicable diseases strain global health systems, prevention of diseases is more important than ever. Existing risk prediction models often lack holistic approaches, and expert opinion remains the gold standard, particularly in certain health topics. However, expert opinion is a scarce and valuable resource. Furthermore, experts are often only consulted when health problems have already come up. Both these problems have a chance to be addressed by having accessible, digital tools available to the general public. However, these can only benefit patients and the healthcare system if they provide valid and actionable information.

The central problem is that “risk” is not a well-defined term when it comes to health – there is no universally accepted definition. Despite this, there are various risk calculator tools available which e.g. assess a user’s risk of developing a cardiovascular disease within the next 10 years or prescribe a “Heart Age”. These are usually very specific and not transparently validated.

This research project aims to understand the current state of risk prediction by evaluating existing risk prediction tools, expert opinion, and a novel, general-purpose digital health risk assessment model called Health Check.

This project will generate insights into the validity of tools by determining whether a higher risk output by the tool would in fact correspond to a shorter time to disease. We will perform an equivalent analysis with physicians where we ask them to order a set of patient profiles according to their risk. As evident from a systematic literature review we are currently conducting, very little research has been published on either the validity of tools or the accuracy of experts.

In conclusion, this research endeavours to advance the understanding of medical risk assessment capabilities and the validity across various disease groups. As a secondary output, this project enables the validation of a digital tool that will enable people to predict and prevent their diseases. The overarching goal is to enable cost-efficient prevention of diseases in the general public, reducing the increasing pressure on the healthcare system and improving the quality of life on an individual level.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comparative-analysis-of-risk-profiles-and-longitudinal-changes-in-risk-factors-for-colorectal-and-lung-cancer-insights-from-the-uk-biobank

Comparative Analysis of Risk Profiles and Longitudinal Changes in Risk Factors for Colorectal and Lung Cancer: Insights from the UK Biobank

Last updated:
ID:
494920
Start date:
17 December 2024
Project status:
Current
Principal investigator:
Dr Raha West
Lead institution:
Imperial College London, Great Britain

This project aims to conduct a comprehensive analysis of risk profiles and longitudinal changes in risk factors for colorectal cancer (CRC) and lung cancer using the UK Biobank data. The study will address critical knowledge gaps by comparing demographic, lifestyle, genetic, and comorbidity risk factors between these two cancers while also assessing how these factors change over time and influence cancer outcomes.

Objectives:

Comparative Analysis of Risk Profiles: We will compare risk factors such as age, sex, ethnicity, smoking status, alcohol consumption, diet, physical activity, comorbidities (e.g., diabetes, cardiovascular disease), and genetic predispositions (e.g., polygenic risk scores) between participants diagnosed with CRC and lung cancer. The study will also explore subgroup differences (e.g., by age, sex, ethnicity) to identify unique and shared risk factors across these cancers.
Longitudinal Analysis of Risk Factor Changes: Using longitudinal data, we will examine how changes in risk factors (e.g., smoking cessation, weight changes, onset or resolution of comorbidities) over time impact cancer outcomes such as survival and recurrence for CRC and lung cancer. This analysis will help identify critical periods where interventions may have the greatest impact.
Methods:

The study will use multivariable logistic regression to identify independent risk factors for each cancer type and Cox proportional hazards models to assess the effect of risk factor changes over time on cancer outcomes. Subgroup analyses will be conducted to explore variations in risk profiles and outcomes based on demographic and genetic factors.

Expected Impact:

The findings will enhance understanding of the differential risk factors and disease mechanisms between CRC and lung cancer, improve cancer risk prediction models, and inform targeted prevention strategies, ultimately contributing to reduced cancer burden and better personalized treatment approaches.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comparative-effectiveness-of-sodium-glucose-cotransporter-2-inhibitors-and-glucagon-like-peptide-1-receptor-agonists-on-cardiorenal-outcomes-in-patients-with-cardiovascular-kidney-metabolic-syndrome

Comparative effectiveness of sodium-glucose cotransporter-2 inhibitors and glucagon-like peptide-1 receptor agonists on cardiorenal outcomes in patients with Cardiovascular-Kidney-Metabolic syndrome

Last updated:
ID:
700503
Start date:
4 September 2025
Project status:
Current
Principal investigator:
Dr Jia Zhou
Lead institution:
Tianjin Chest Hospital., China

Cardiovascular-kidney-metabolic (CKM) syndrome is a novel concept that connects cardiovascular diseases, chronic kidney disease and diabetes. Scientific statement from the American Heart Association provides a CKM staging construct for prevention and care optimization within CKM syndrome from stage 0 (no CKM risk factors) to stage 4 (clinical CVD in CKM syndrome) and emphasized the optimal strategies for supporting lifestyle modification, targeting of emerging cardioprotective and kidney-protective therapies.
The cardioprotective therapies of sodium-glucose cotransporter-2 inhibitors (SGLT2is) and glucagon-like peptide-1 receptor agonists (GLP-1RAs) have revolutionized preventive care for individuals with diabetes. SGLT2is, originally developed as antidiabetic agents, are now known to prevent kidney failure and to have cardioprotective effects, most notably on heart failure-related hospitalizations and cardiovascular mortality. GLP-1RAs not only improve insulin resistance and glycemia but also reduce weight and cause significant reductions in the risk of major adverse cardiovascular events (MACE).
However, strategies for prioritizing the selection of SGLT2is or GLP-1RAs in the CKM syndrome are not well defined. Further data are urgently needed to guide prioritization of antihyperglycemic agents in patients with different stages of CKM syndrome.
Although recent studies have compared the effectiveness of SGLT2is versus GLP-1RAs on cardiovascular outcomes in patients with diabetes by emulating target trial, the effectiveness of these two drugs on cardiorenal outcome in different stages of CKM syndrome is lacking. Therefore, the aim of the project is to perform a target trial emulating to compare the effectiveness of SGLT2is versus GLP-1RAs on major adverse cardiovascular events and serious renal events in patients with CKM syndrome.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comparative-genetic-and-analysis-of-pulmonary-diseases-across-different-races

Comparative genetic and analysis of pulmonary diseases across different races

Last updated:
ID:
56719
Start date:
6 January 2020
Project status:
Current
Principal investigator:
Dr Zhu Zhang
Lead institution:
Peking Union Medical College, China

Most of the pulmonary diseases may be influenced by race, ethnicity and other genetic factors. However, limited data exist that compare these pulmonary diseases such as COPD, IPF, VTE et al between different ethnic groups, especially in the Chinese population and Western population. Asia is the most populous continent, accounting for 60% of the world population. As the world’s most populated country, the Chinese population is accounted for like 1/5 of the world’s population. Meanwhile, the evidence shows pulmonary diseases have been the primary diseases affecting Chinese population health. But these patients are always underrepresented in human genetics research. Our research results will add important evidence on the genetic diversity in Chinese patients and UK patients in pulmonary diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comparative-risk-evaluation-of-clinical-phenotypes-and-genetic-predisposition-for-end-stage-renal-diseases-and-emotional-vulnerability-across-various-ethnic-populations

Comparative risk evaluation of clinical phenotypes and genetic predisposition for end-stage renal diseases and emotional vulnerability across various ethnic populations

Last updated:
ID:
81803
Start date:
30 March 2022
Project status:
Current
Principal investigator:
Dr Hung-Lin Chen
Lead institution:
China Medical University Hospital, Taiwan, Province of China

The aims of our study are to evaluate the risk of outcomes associated with mental illness among patients with chronic kidney disease (CKD) and end-stage renal disease (ESRD). We would also evaluate the burden of CKD among patients with mental disorders.

Mental disorders are associated with worse survival in CKD/ESRD patients. CKD /ESRD patients have been observed with anxiety and cognitive-dysfunction. In addition, people with severe mental illness have less chance to receive routine CKD screening. Therefore, there has been less work on the effect of psychiatric illness on CKD/ESRD prognosis and survival. A subset of CKD/ESRD patients often have difficulties with multitasking, short-term memory, word-finding, and attention. The underlying mechanism is unclear but may include direct neurotoxic effects of renal replacement therapy, oxidative damage, and genetic predisposition. Lifestyle and environment are also important parameters as potential risk factors. Conversely, the epidemiology of CKD/ESRD among patients with mental disorders remains to be evaluated. More research is required to clarify the issue of CKD care access equality.

We propose to utilize data from different populations in 3 years to examine the associations between mental illness and CKD/ESRD. The first year will be utilized to examine the factors associated with developing CKD/ESRD for patients with mental issues and to check for risk stratification effect due to sex, age and race for the full spectrum of CKD/ESRD. Also demographic characteristics, health status, lifestyle effects and CKD/ESRD risk factors will be compared for different mental health subgroups. Furthermore, genomic data will be used to conduct association tests to detect significantly associated single nucleotide polymorphisms (SNPs), copy number variations (CNVs), and for genetic correlations amongst different CKD/ESRD and mood-related conditions. The second year will be used to conduct survival analysis for patients with respect to different demographic, clinical and genomic factors. Also significantly associated findings from first year and imaging data would be used to train deep-learning models to predict prognosis and survival of CDK/ESRD. Finally, in the third year, comparison studies will be done between UK biobank and Taiwanese samples to establish ethnicity specific findings.

Psychiatric patients are less aware of CKD/ESRD but likely die of it. The absence of adequate data makes it difficult to predict mental health impact on CKD/ESRD patients. This study will establish factors that affect the emotional vulnerability and comorbidity of mental health and ESRD among patients and providing necessary information that has been lacking in this field.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comparing-genome-architecture-across-diverse-populations-between-the-uk-biobank-and-the-biobank-at-the-colorado-center-for-personalized-medicine-ccpm

Comparing genome architecture across diverse populations between the UK Biobank and the Biobank at the Colorado Center for Personalized Medicine (CCPM)

Last updated:
ID:
100578
Start date:
26 April 2023
Project status:
Current
Principal investigator:
Dr Joanne Cole
Lead institution:
University of Colorado, Anschutz Medical Campus, United States of America

The Colorado Center for Personalized Medicine (CCPM) aims to advance the health of individuals, families, and communities by integrating multi-omics with detailed participant information. Established in 2014, CCPM, together with the University of Colorado School of Medicine and the local healthcare system UCHealth, brings together health records from our diverse region with a biospecimen repository for research, clinical, and educational use. As a genome-centered team, we focus on mapping genetic and environmental determinants of health and disease, developing and improving statistical approaches to large-scale multivariable and multi-omic data, harnessing genomics to inform our understanding of human diversity, and predicting disease risk and health outcomes to improve clinical care. Together with our local biobank, we will utilize the complementary prospective and in-depth design of UK Biobank and its full dataset to support our joint missions to advance modern medicine and treatment through scientific discovery. Specifically, we will identify novel determinants of human health, develop methods to improve multi-omic analysis, compare statistical association results across disease and trait domains from genetic ancestry-matched or self-identified race/ethnic groups as appropriate, and importantly share approaches and discoveries with UK Biobank, the scientific community, and the general public for collective and global human health advancement. Importantly, contrasting the two datasets gives us an opportunity to understand differences in healthcare utilization or access patterns across both countries.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comparing-polygenic-risk-and-predicted-loss-of-function-plof-variants-in-a-healthy-ageing-cohort-aspree-with-the-uk-biobank-across-a-range-of-disease-and-health-related-phenotypes

Comparing polygenic risk and predicted loss-of-function (pLoF) variants in a healthy ageing cohort (ASPREE) with the UK Biobank, across a range of disease and health-related phenotypes

Last updated:
ID:
47061
Start date:
12 March 2019
Project status:
Current
Principal investigator:
Professor Paul Lacaze
Lead institution:
Monash University, Australia

Our research aims to determine if genetic risk factors seen in the UK Biobank are the same as those seen in a healthy elderly population from Australia (the ASPREE cohort). In particular, we’d like to test whether there are differences in the frequency or severity of known genetic risk factors between the two populations. We would also like to test for protective genetic factors in the healthy elderly, which might off-set disease risk.
Aim 1: To compare the distribution and frequency of common genetic risk factors (inferring risk of common disease) between the UK Biobank and the ASPREE cohort.
Aim 2: To compare the frequency, distribution and medical relevance of rare, damaging genetic risk factors between the populations, and determine if they are depleted in the healthy elderly across a range of disease outcomes.
Aim 3: To compare the frequency, distribution and medical relevance of protective genetic factors in the healthy elderly.
This project is expected to last for many years, as we plan to continue to monitor the health of the ASPREE healthy elderly population for another 5 years, or more.
The public health impact of our research includes improving the understanding of human genetic variation, helping understand how genetic factors contribute to disease risk, and helping understand how genetic factors may contribute to positive health outcomes, such as healthy ageing. This is particularly relevant given the growing burden of ageing populations in many countries around the world, and the uncertainty with regards to the contribution of genetic versus environmental factors.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comparing-resting-state-networks-in-post-traumatic-stress-disorder-ptsd-in-cannabis-and-non-cannabis-users-and-healthy-controls-in-uk-biobank-sample

Comparing resting state networks in post-traumatic stress disorder (PTSD) in cannabis and non-cannabis users and healthy controls in UK Biobank sample

Last updated:
ID:
96894
Start date:
6 March 2023
Project status:
Current
Principal investigator:
Dr Arpan Dutta
Lead institution:
University of Manchester, Great Britain

Aims:
The aim of this project is to determine the difference in resting brain networks in those who have have PTSD between those who use cannabis and do not use cannabis

Rationale:
Previous neuroimaging research has revealed there are differences in brain structure and activity in people who have PTSD and in people who use cannabis. Cannabis based medicines are currently being researched as a potential treatment for PTSD. Using MR imaging techniques we can look at how cannabis usage affects the functioning of interconnected brain areas in PTSD. Presently, there is no research on resting state imaging comparing people who have PTSD and use cannabis and those who have PTSD but do not use cannabis. Therefore, this project will provide useful information in this current gap of knowledge to aid research and understanding about the effects of cannabis on the brains of people who have PTSD.

Project duration:
This project will be completed within 3 years
Public health impact:
As cannabinoids are being researched as a potential treatment for PTSD, this research will have an impact on understanding in this area. The results can be used to inform future research on cannabis use in PTSD and the effects this may induce on brain structure and activity.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comparing-variant-distributions-and-polygenic-risk-score-between-the-uk-biobank-and-south-asian-cohorts

Comparing variant distributions and Polygenic Risk Score between the UK Biobank and South Asian cohorts

Last updated:
ID:
100235
Start date:
5 May 2023
Project status:
Current
Principal investigator:
Dr Radhakrishna Bettadapura
Lead institution:
Strand Life Sciences Pvt Ltd, India

As part of our project codenamed PhenoGen, we are collecting deep phenotypic and genomic data on individuals from Central and South India. We plan to jointly analyze this cohort with the UKBB cohort to understand the variant distributions between these two cohorts and to identify novel disease-specific genetic variants that can provide new insights into disease biology. One-fourth of the global population is of South Asian ancestry, but the representation of South Asian genomes in global genomic studies remains sparse, with a major focus being on Caucasian individuals. By expanding the South Asian cohort substantially using the PhenoGen cohort (phase 1: 10K, phase 2: 100K), we aim to get a deeper understanding of how genomic variants in the South Asians impact various diseases of interest. We also propose to build risk predictors for NAFLD/NASH and CAD with better applicability to South Asians.

NAFLD/NASH and CAD have high prevalence in India and risk prediction and new therapeutics for both areas are of great societal interest. NAFLD/NASH is the leading cause of liver-related mortality with an ever increasing prevalence rate and no approved pharmacotherapy till date. Similarly, the incidence, prevalence, and mortality from premature CAD in Indians and other South Asians is reportedly ~50-400 times higher as compared to any other ethnic group. Both NAFLD/NASH and CAD are known to have significant genetic underpinnings, and also have a high heritable component. With the PhenoGen project, we hope to expand our understanding of the genetic architecture of both these diseases by deepening the knowledge of the molecular mechanisms, pathophysiological processes and the interplay between genetic factors for the identification of novel therapeutic targets. Our initial focus is on these two areas, but we hope to look at other disease areas in due course of time.

Given that disease risk stems from a combination of rare and common genomic variants, we propose to analyze the UKBB cohort and the PhenoGen cohorts jointly to identify rare and common variant loci of interest to the above two disease areas that could provide better ways to predict risk as well as yield new drug targets. In the process, we would also like to do a broader variant distribution analysis across the two cohorts. We expect the project to start in Jan 2023 and continue for 2 years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comparison-and-replication-of-gwas-and-gene-environment-interaction-signals-and-polygenic-risk-scores-for-complex-traits-in-the-african-awi-gen-cohort-study

Comparison and replication of GWAS and gene-environment interaction signals and polygenic risk scores for complex traits in the African AWI-Gen cohort study

Last updated:
ID:
63215
Start date:
26 November 2020
Project status:
Current
Principal investigator:
Professor Michele Ramsay
Lead institution:
University of the Witwatersrand, Johannesburg, South Africa

African populations are experiencing an increase in the health burden of non-communicable disease, as has been experienced in developed regions of the world over the past 50 years. The sustainability development goals have therefore emphasized the need to reduce the prevalence of obesity, high blood pressure, diabetes, cancer and lung disease, among many others. The aim of our study is the compare and replicate the outcomes from genome-wide association studies (GWASs) and gene-environment interactions, as well as polygenic risk scores, detected or developed in the African AWI-Gen cohort study to data from the UK Biobank. AWI-Gen is an African population-based cohort of ~12,000 male and female participants (40 to years old at baseline) from Ghana, Burkina Faso, Kenya and South Africa with data similar to, though not as extensive as that in the UK Biobank. There are few cross-sectional population studies of older adults in Africa making it difficult to do replication studies to test the robustness and transferability of results. We will use the UK Biobank to strengthen our studies and to better understand the similarities and differences in the contributions of genetic susceptibility and gene-environment interaction to disease between African and non-African populations. Since Africa is the cradle of humankind and has genetic variants not found elsewhere in the world, there is an opportunity to make novel discoveries that could inform improved treatments and health outcomes worldwide.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comparison-of-accelerometer-derived-physical-activity-patterns-among-current-cancer-patients-cancer-survivors-and-cancer-free-population

Comparison of accelerometer-derived physical activity patterns among current cancer patients, cancer survivors, and cancer-free population.

Last updated:
ID:
44513
Start date:
21 November 2018
Project status:
Closed
Principal investigator:
Mr Mustafa Oguz
Lead institution:
Evidera, Inc., Great Britain

The number of patients currently living with or has survived cancer in the United Kingdom is estimated at around three million. Each year there are an estimated 360,000 new cancer diagnoses. Studies that investigate the relationship between physical activity (PA) and cancer suggest that PA is an important predictor of outcomes prior to, during, and after a cancer episode. Examples of findings include lower risk of cancer for high levels of leisure-time PA, and reduced cancer related fatigue for breast and prostate cancer patients who joined an exercise intervention.
In accordance with the literature on the potential benefits of PA to achieve better health outcomes for cancer patients, guidelines for cancer patients recommend promotion of PA carefully tailored to the individual at all stages of cancer.
However, cancer patients and survivors are more likely to be inactive compared to cancer-free population. A comparison of PA levels of cancer patients, survivors, and cancer-free population can help establish the deterioration of PA levels among patients with cancer and rehabilitative needs of cancer survivors. UK Biobank’s baseline and repeat assessments, accelerometer study, and links to cancer registries for a large number of participants has the potential to provide insight into these comparisons.
We therefore aim to answer three research questions:
! Do accelerometer-derived physical activity patterns differ between current cancer patients and the cancer-free population?
! Do accelerometer-derived physical activity patterns differ between cancer survivors and cancer-free population?
! Are physical activity patterns predictive of cancer diagnosis among cancer-free population?
We will first compare the physical activity patterns between current cancer patients and cancer-free population, then compare the physical activity patterns between cancer survivors and the cancer-free population. To answer the last question, we will start with UK Biobank participants who never had a diagnosis of cancer at the time of accelerometer study, and estimate how the risk of cancer diagnosis changes with PA patterns by looking at cancer diagnoses these participants might later have.
This research will help establish the physical activity levels of cancer patients and survivors in the UK, highlight gaps compared to the general population. The study can guide physical activity interventions towards specific cancer types. The identification of associations between physical activity levels and cancer incidence from a large study such as UK Biobank will be an important contribution to the literature on this topic.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comparison-of-computational-and-statistical-methods-for-identifying-genetic-risk-factors-for-cognitive-decline-in-parkinsons-disease

Comparison of computational and statistical methods for identifying genetic risk factors for cognitive decline in Parkinson’s Disease

Last updated:
ID:
67829
Start date:
16 June 2021
Project status:
Current
Principal investigator:
Professor Holger Fröhlich
Lead institution:
Fraunhofer Institute, Germany

Aim: Aim of the project is to identify genes associated to cognitive decline in Parkinson’s Disease.
Scientific rationale: Idiopathic Parkinson’s Disease (PD) is influenced by genetic variants. More specifically, there is likely a genetic contribution to the level of cognitive decline, which is frequently observed in PD patients. However, identifying corresponding genetic variants via classical statistical approaches remains challenging, specifically in case of rare genetic variants. Hence, statistical and computational approaches are of interest that aggregate variants, e.g. on gene level. A number of methods have been proposed, but we need to better understand their advantages and limitations, including a systematic power analysis.
Following such an analysis we will apply the most computational promising approach to unravel genes associated with cognitive decline in PD. The knowledge of such genes is an important step towards finding new and better medications in the future.
Project duration: 1 year
Public health impact: This project focuses on identifying genetic factors that contribute to cognitive decline in Parkinson’s Disease (PD). Identifying such genetic factors is important to develop novel therapies in the future.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comparison-of-different-genetic-modeling-methods-for-complex-genetic-conditions

Comparison of different genetic modeling methods for complex genetic conditions

Last updated:
ID:
34802
Start date:
3 September 2018
Project status:
Closed
Principal investigator:
Mr Bernard Stopak
Lead institution:
Ada Health GmbH, Germany

There is a currently a large focus in patient-facing genomics to accurately identify individuals who are at risk for heritable or partially heritable diseases. To-date, there have been advances in accounting for this heritable risk, but gaps in performance remain as there remains a portion of unexplained inheritance. Our goal is to produce genetic risk prediction models that can accurately assess whether an individual is at an increased risk for disease. We believe that complex phenotypes have differing underlying genetic architecture. Therefore, it may be that different modeling methods will work better for different conditions. For example, for a condition with several high-effect variants, such as age-related macular degeneration, a simple genetic model may perform well with few genetic variants. However, for a condition like Crohn’s disease where hundreds of genetic loci have been identified, more complex models with more genetic variants are likely to be more powerful. We will create and test our genetic predisposition models for their validity to predict individuals at-risk of genetically correlated diseases using polygenic risk scores, machine learning, and hand-curated genetic models. These models will then be compared to see which kind of modeling best reflects the genetic architecture of each condition, then see if there are any patterns amongst different conditions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comparison-of-hla-imputed-genotype-of-the-general-population-versus-population-with-incidence-of-cancer

Comparison of HLA imputed genotype of the general population versus population with incidence of cancer.

Last updated:
ID:
53781
Start date:
15 January 2020
Project status:
Closed
Principal investigator:
Dr Robert Bentham
Lead institution:
University College London, Great Britain

What determines whether someone develops cancer or not? Over 36 months we aim to investigate the role of the immune system in this process.

The immune system can recognise and kill cancer cells. To propagate and grow, cancer has to escape this recognition using different mechanisms. Cancer is well known to vary incredibly between patients as well as within an individual tumour, but the immune system however is also incredibly genetically varied. How someone’s immune system functions is largely determined by three genes HLA-A, HLA-B and HLA-C. Of these three genes there are thousands of different variations known as alleles, and with there being two alleles of each gene in each person genomes the ‘HLA type’ of any two non-related people is likely to be different.

Knowing a patient’s HLA type is important for many new types of cancer treatment including immunotherapy. It is not well known however how exactly a person’s HLA type determines the general course and evolution of cancer as a disease. We hypothesise that there may be some combinations of these HLA genes that are more able to recognise and kill cancer types leading to a lower incidence of cancer in this population.

Part of the challenge of answering this question is that there are so many different HLA types possible. By using a large dataset such as the UK biobank and grouping similar HLA types together we hope to overcome this and if a person’s HLA type can be shown to be predictive of cancer risk it will provide a very useful tool enabling increased monitoring of high risk populations.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comparison-of-lipid-derived-indices-with-conventional-lipoprotein-tests-and-apolipoproteins-for-cardiovascular-disease-prediction-using-uk-biobank-data

Comparison of Lipid-Derived Indices with Conventional Lipoprotein Tests and Apolipoproteins for Cardiovascular Disease Prediction Using UK Biobank Data

Last updated:
ID:
531547
Start date:
4 December 2024
Project status:
Current
Principal investigator:
Dr Bin Wang
Lead institution:
Beijing Tsinghua Changgung Hospital, China

The objective of this project is to compare various lipid-derived indices with conventional lipoprotein tests and apolipoproteins to identify the most valuable biomarkers for cardiovascular disease (CVD) prediction. Using data from the UK Biobank, we will assess a range of lipid measurements, including total cholesterol, HDL-C, non-HDL-C, direct and calculated LDL-C, apolipoproteins A1 and B, and novel lipid-derived markers. Through Cox proportional hazard models adjusted for classical CVD risk factors, we will evaluate the predictive value of these lipid indices in association with both fatal and nonfatal CVD events. The study aims to determine whether novel lipid-derived markers offer superior predictive accuracy compared to conventional lipid profiles and apolipoproteins. Our goal is to identify the most effective biomarkers for CVD risk assessment, providing evidence to guide future clinical practices in cardiovascular risk evaluation.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comparison-of-obesity-paradox-in-diabetic-and-nondiabetic-population

Comparison of `obesity paradox` in diabetic and nondiabetic population

Last updated:
ID:
18815
Start date:
1 September 2016
Project status:
Closed
Principal investigator:
Professor Thomas Yates
Lead institution:
University of Leicester, Great Britain

Current evidence on the association between BMI and mortality both in
the general population and diabetics is inconclusive with some studies
reporting a linear association, some reporting a U shaped curve and some
attributing the shape of the effect on factors like age, sex, ethnicity etc.
Therefore, we aim to:
-assess the association between BMI and mortality in the general
population and diabetic population
– investigate whether these associations differ across several potential
effect-modifiers (eg, sex, ethnicity, physical activity, smoking) The UK Biobank is aimed at supporting research intended to improve prevention, diagnosis, and treatment of illness and promotion of health. This project aligns very closely with the purpose of Biobank and addresses a very important issue in the current health care settings in the UK. A better understanding of the association between body mass index and mortality in both diabetic and nondiabetic populations will not only feed into the recommendations for the management of diabetes and other chronic illnesses but also shed light on whether these recommendations need to be different for different groups (smokers,ethnic groups,males/females) We will quantify the risk of death associated with different body mass index (BMI) in diabetics and non-diabetics to assess whether the shapes of association are different. Furthermore, we will also assess whether the risk of death associated with different levels of BMI differs by patient factors (e.g. ethnicity,age, gender, smoking status, physical activity, physical fitness etc.) Full cohort


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comparison-of-the-activity-and-expression-of-the-bace-family-of-proteins-with-genetic-splice-variants-and-the-associations-with-disease

Comparison of the activity and expression of the BACE family of proteins with genetic/splice variants, and the associations with disease.

Last updated:
ID:
102416
Start date:
26 April 2023
Project status:
Current
Principal investigator:
Dr Paul Meakin
Lead institution:
University of Leeds, Great Britain

A hallmark of Alzheimer’s Disease are plaques in the brain, which are made up of small fragments of protein called amyloid beta peptides. Amyloid beta peptides are produced via cutting of the amyloid precursor protein (APP) by beta secretase 1 (BACE1). BACE1 activity is increased in Alzheimer’s Disease, and is therefore pivotal to the development of the disease. Research on the role of BACE1 outside of Alzheimer’s Disease is limited. Recently, increased BACE1 activity has been implicated in other functions and diseases, including diabetes and obesity. BACE1 has been a therapeutic target of interest over the past couple of decades. However, to date there has not yet been any success, with clinical trials ending due to no improvement or worsening of cognitive function, or due to safety concerns. Several BACE1 inhibitors have been reported to also target BACE2, some to a greater extent than BACE1. BACE2 is a family member of BACE1, known to compete for the same substrates. The wide-ranging reported side effects of these drugs highlight the limits in understanding of the roles of BACE1 and BACE2.

We believe that BACE1 and BACE2 have a range of functions that are not yet understood, and may be implicated in other diseases. Recently BACE1 has been linked to obesity, diabetes mellitus, cancer and cardiovascular disease. We will use a wide range of UK Biobank data to look at whether changes in BACE1 and BACE2 activity are related to genetic variations between people. The amount of data available on UK Biobank will then allow us the excellent opportunity to study BACE1 and BACE2 relevance to many diseases. This may shed light on new roles of these enzymes in disease, and due to the existence of BACE1 inhibitors, provide novel uses for these drugs.

This project will be undertaken over approximately 3 years, contributing towards a PhD project. We hope the findings will shed light on the role of BACE1 and BACE2 in disease, and potential mechanisms behind the increases in BACE1 activity observed in various diseases. Ultimately, we hope that it may lead to novel targets for BACE1 inhibitors, which could have a significant impact on the treatment of disease, and a potential for a personalised medicine approach.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comparison-of-the-association-of-fat-mass-bmi-and-other-obesity-indicators-with-all-cause-and-cause-specific-mortality

Comparison of the association of fat mass, BMI, and other obesity indicators with all-cause and cause-specific mortality

Last updated:
ID:
59776
Start date:
21 September 2020
Project status:
Closed
Principal investigator:
Dr Zhenhua Xing
Lead institution:
Central South University, China

Aim: we evaluate the effectiveness of fat mass, BMI, and other obesity indicators(waist circumference, Waist-to-Hip ratio)to predict all-cause and cause-specific mortality
scientific rationale: it is still unclear which indicator of obesity is highly close to all-cause mortality and cause-specific mortality.
public health impact: The founding of our study will help us control obesity and choose suitable indicators of obesity
project duration:3 years


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comparison-of-the-genetic-architecture-between-latino-and-ukbb-populations-as-strategy-to-understand-its-role-on-human-health-traits-and-diseases

Comparison of the genetic architecture between Latino and UKBB populations as strategy to understand its role on human health traits and diseases.

Last updated:
ID:
89006
Start date:
25 August 2022
Project status:
Current
Principal investigator:
Dr Carlos D Bustamante
Lead institution:
Galatea Bio, inc, United States of America

The lack of representation of non-European individuals on genetic studies introduces a major bias on health. The number of studies for admixed groups, i.e. Latino populations, represents less than 2% out of the total number of medical genomics studies in the field.
This proposal aims to compare the genetic architecture of the UKBB biobank participants, mostly composed of white British individuals, with admixed individuals from Latino groups, and quantify its impact in three selected medical areas of high impact at the community level, cardiometabolics traits and diseases, cancer and Covid-19 related phenotypes.
We plan to conduct a three-year project that led us: 1) to deeper explore the genomic population structure of UKBB population, and admixed groups; 2) to identify new genetic variants associated with cardiometabolics traits and diseases, cancer and Covid-19 related phenotypes; and 3) to estimate the genetic risk scores for admixed groups considering prior findings on the UKBB dataset.
This effort might facilitate the discovery of new genetic findings, that guide prevention, treatment, and monitoring efforts for admixed individuals; helping to improve health at multi-ethnic level.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comparisons-of-data-collected-on-women-participating-in-both-uk-biobank-and-the-million-women-study

Comparisons of data collected on women participating in both UK Biobank and the Million Women Study

Last updated:
ID:
78571
Start date:
27 June 2022
Project status:
Current
Principal investigator:
Dr Sarah Floud
Lead institution:
University of Oxford, Great Britain

The aim of this project is to enable comparisons to be made between data collected in the Million Women Study and UK Biobank. About 50,000 Million Women Study participants have also participated in the UK Biobank study, and the information that participants have given in the two studies can be compared for accuracy and reliability, as well as assessing changes over time. Strict data protection controls will be applied and our comparisons will use a dataset with identifying information removed. The Million Women Study has obtained ethical approval for the linkage. Over the course of the project (initially 3 years duration), different measures will be compared. This will enable more reliable evidence on lifestyle risk factors for major chronic diseases to be provided for the benefit of public health.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comparisson-of-smoking-behaviour-between-men-and-women-to-assess-risk-inducing-habits

Comparisson of smoking behaviour between men and women to assess risk inducing habits

Last updated:
ID:
2482
Start date:
1 October 2013
Project status:
Closed
Principal investigator:
Professor Mark Woodward
Lead institution:
George Institute for Global Health, Australia

We have recently completed meta-analyses involving millions of subjects, which showed that, although smoking increased risk considerably in both sexes, women who smoke have a 25% greater excess relative risk for coronary heart disease compared to men who smoke. Although not as strong, there is also a trend towards a similar excess risk for stroke among women smokers compared with men. Furthermore, we have published other data, which suggest an excess relative risk of around 100% from smoking for women, compared to men, for lung cancer mortality. One possible reason for these excess relative risks in women is that their smoking habits tend to be more risk-inducing. For example, they may smoke more, or start at a younger age. On the other hand, it may be that women have less risk-inducing smoking habits, in which case the excess relative risks we, and others, have found are even more remarkable, with implications for targeted public health measures to prevent smoking and promote quitting. We request the use of the baseline Biobank data on smoking habits to make comparisons between women and men, so as to understand whether variations in habits may explain the excess relative risks we have found. We shall also explore whether sex differences persist within important subgroups of the Biobank population, by age, self-reported illness, socio-demographic status, and ethnicity. These will all be cross-sectional analyses; in the future we propose to make a further application to investigate whether sex-specific smoking habits contribute to the risk of the major smoking-related chronic diseases using longitudinal Biobank data.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comparissons-of-measures-of-adiposity-including-body-mass-index-waist-circumference-waist-to-hip-ratio-and-percentage-body-fat-mass

Comparissons of measures of adiposity including body-mass index, waist-circumference, waist-to-hip ratio, and percentage body fat mass

Last updated:
ID:
7439
Start date:
1 September 2013
Project status:
Current
Principal investigator:
Professor Emanuele Di Angelantonio
Lead institution:
University of Cambridge, Great Britain

There has been a dramatic increase in adiposity over the last few decades, resulting in more than 1 billion overweight adults and 300 million obese worldwide. Excess body fat has been associated with several biochemical, lifestyle and other characteristics, as well as with multiple chronic diseases, including type 2 diabetes, coronary disease, stroke, and several site-specific cancers.

However, previous studies have been underpowered to directly compare these associations across various measures of adiposity, including body-mass index, waist-circumference, waist-to-hip ratio, and percentage body fat mass.

The objective of this research is:

(1) to assess precisely any cross-sectional associations of adiposity measures with biological, lifestyle and other characteristics (including biochemistry markers, when available). This analysis will help to (i) determine to what extent adiposity measures share related information; (ii) investigate the determinants of adiposity measures; and (iii) investigate potential biological pathways of the underlying association between adiposity and disease.

(2) to determine within-person variability in adiposity measures using serial measurements.

(3) to characterise and compare the associations of adiposity measures with future risk of all-cause mortality, and, when sufficient events/deaths are available in UK Biobank, with risk of site-specific cancers and cause-specific mortality.

This research involves the use of data only (ie, no samples are required) and will help to better understand factors that affect adiposity levels and clarify the relative importance of adiposity measures on disease risk. The number and the range of risk factors available in UK Biobank provide a unique opportunity to study such associations in all participants with information on weight, height, waist and hip circumference, and percentage body fat. In the first phase, we would also require information on socio-demographic, lifestyle, environment, early life, psychosocial and physical measures. When available in UK Biobank, we would like data on biomarkers and serial measurements of adiposity measures to investigate their within-person variability.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comperehensive-meta-analysis-of-the-clinical-and-molecular-features-of-clonal-hematopoiesis

Comperehensive meta-analysis of the clinical and molecular features of clonal hematopoiesis.

Last updated:
ID:
213770
Start date:
6 March 2025
Project status:
Current
Principal investigator:
Dr Ryunosuke Saiki
Lead institution:
Kyoto University, Japan

Recent advances in genetic testing have revealed that clonal hematopoiesis (CH)-a condition that can lead to blood cancers-also appears in healthy individuals and is linked to higher risks of many diseases. However, further analysis is needed to understand how different types of CH, each with specific genetic changes, relate to these disease risks. Additionally, the biological reasons behind these links remain unclear. Although some genetic and environmental factors associated with CH have been identified in European populations, they have not been studied in Asian populations, leaving regional differences unknown. Addressing these issues could enhance our understanding of CH and its potential clinical applications. In this project, we will analyze genetic and clinical datasets from the UK Biobank (UKBB) and other cohorts to address the following questions:
How accurately can disease risk be predicted based on CH characteristics?
What are the biological mechanisms linking CH to various diseases?
Which genetic and environmental factors contribute to the development of CH?
How do CH characteristics differ across ethnic groups?
The expected duration of this project is three years.
Our analysis will clarify the clinical and biological traits of CH in detail, helping to assess disease risks by CH subtype and guide risk management. By studying CH’s molecular features, we aim to understand its connection to disease risks and identify new treatment targets.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/complementing-human-data-centric-approaches-to-discover-novel-therapies-and-biomarkers-at-novo-nordisk

Complementing human data centric approaches to discover novel therapies and biomarkers at Novo Nordisk

Last updated:
ID:
86252
Start date:
14 September 2022
Project status:
Current
Principal investigator:
Dr Dmitry Shungin
Lead institution:
Novo Nordisk A/S, Denmark

Despite tremendous advances in contemporary biomedical research, the success rates of drug discovery from initial idea to approved medicines remain low resulting in substantial financial burden on healthcare systems around the globe. Large-scale population-level datasets with genetic and other omics data layers such as UK Biobank are one of the key resources of real-world human evidence that could improve and accelerate the process of new drug discovery. Such collections of data open new opportunities both to personalize medicines by finding sub-populations of individuals with favourable treatment response; as well as to efficiently estimate drug efficacy and safety at early stages of drug development process. A rich panorama of phenotypes available in UK Biobank, such as imaging, can further strengthen attempts to search for new therapies when combined with omics data.
Thus, the aim of this proposal is to utilize multiple layers of data from UK Biobank to potentiate and complement novel target and biomarker discovery efforts in Novo Nordisk with focus on multiple therapeutic areas, including but not limited to obesity, diabetes, cardiovascular, kidney and liver diseases.
To achieve that aim we will utilize all available UK Biobank data to identify new and re-purpose existing targets and biomarkers within Novo Nordisk disease portfolio. Using state of the art statistical and bioinformatic methods we will connect and cross-reference individual genetic profiles, clinical markers, metabolomics and proteomics readouts, imaging data, as well as other available data points to obtain the most extensive evidence linking particular drug targets with relevant diseases at multiple levels of biological comprehension. This systematic approach will allow for faster drug development cycles, as well as will streamline spending of resources by prioritizing therapies that have the highest potential to help people suffering from key chronic conditions.
Using plethora of UK Biobank data, we will also strive to identify sub-groups of individuals with differences in disease features or treatment responses which will help us to personalize therapies for people who benefit the most and guide our clinical trials to be more personalized, with less risk to patients and faster trial completion as a result.
Thus, evidence from UK Biobank combined with other resources in Novo Nordisk will galvanize development of novel prevention strategies and innovative therapies that will improve and prolong lives of patients burdened with chronic conditions around the globe. This proposal therefore fits well with the UK Biobank’s purpose of improving the prevention and treatment of illness.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/complementing-rare-disease-studies-with-data-from-national-cohort-projects

Complementing rare disease studies with data from national cohort projects.

Last updated:
ID:
63533
Start date:
27 August 2020
Project status:
Current
Principal investigator:
Professor Amy Jayne McKnight
Lead institution:
Queen's University Belfast, Great Britain

Approximately six percent of the UK population are affected by a rare disease at some point in their lives. Rare diseases are often associated with a long diagnostic odyssey, significant annual healthcare costs, few effective treatment options, and major impacts on day-to-day living for patients and their families. An accurate diagnosis and early intervention may significantly improve patient outcomes.

We plan to combine the rich resources available within UK Biobank with other population-based datasets to improve patient focused clinical care. Our group is keen to discover molecular features that influence rare diseases; we will take a multi-omic approach to improve diagnosis and identify molecular features that influence rare disease pathways. We are also keen to improve support for people with rare diseases, who often experience significant social isolation. UK Biobank provides a very rich resource to compare access to services, pain, activity, loneliness, and support experienced by people with a rare disease compared to common diseases, compared to a group of people of a similar age who are unaffected by these conditions.

This study will not directly help UK Biobank participants, but will help deliver robust research to aid clinical decision making and diagnosis, with the hope this could lead to improved outcomes for people with rare diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/complete-association-analysis-of-inversions-and-other-complex-genomic-variants-with-human-diseases-and-health-related-traits

Complete association analysis of inversions and other complex genomic variants with human diseases and health related traits

Last updated:
ID:
352687
Start date:
29 January 2025
Project status:
Current
Principal investigator:
Dr Mario Caceres Aguilar
Lead institution:
Hospital del Mar Medical Research Institute, Spain

An important aim of current biomedical research is finding the genetic basis of individual characteristics. Since the development of the human genome project, multiple studies have created deeper catalogues of human genetic variants. However, not all the classes of variants have been studied to the same level of detail. Inversions are one type of genetic variants that affect a large fraction of the human genome and that have been implicated in differences between individuals, both in humans and other organisms. Nevertheless, they have been poorly studied due to technical challenges in their detection, which has precluded determining their role in different diseases and other traits of interest. In addition, inversions are usually located in complex genomic regions that include several rearrangements and that are very difficult to characterize with current methods. In this project, we will take advantage of all the knowledge generated about these variants during the last years to fill an important void in the study of the genetic basis of human diseases. Specifically, we will use different computational methods to infer accurately as many as possible of these little-known variants in the UK Biobank population. Next, we will use the extremely useful information available of each individual to investigate their role in the susceptibility of multiple common diseases and other health-related traits. Thus, this analysis could uncover previously unknown disease associations of inversions and complex variants, which could result in potential improvements in diagnostics and therapy with significant social benefits. Moreover, we will generate new data that could be used in future studies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/complex-analysis-and-creation-of-extensible-datasets-for-ukbiobank

Complex Analysis and Creation of Extensible Datasets for UKBioBank

Last updated:
ID:
82561
Start date:
7 June 2022
Project status:
Current
Principal investigator:
Dr Loren Buhle
Lead institution:
DNAnexus, Inc., United States of America

Aims: We are going to make it easier to access and subset on different data types, so researchers can get to meaningful results faster. Scientific Rationale: For precision medicine to be realized, researchers must be able to subset cohorts of patients on imaging, medical test (e.g. electrocardiogram), activity, or metabolic data. Once these subsets are established, treatment options become more precise due to an understanding the genetic contributions to these cohorts’ disease. Public health impact: This subsetting is important because it is often used to direct the treatment protocol for this disease. If the subsetting is done incorrectly and inappropriate drugs are used for each subset, patients may die faster. Project duration: We expect this project to last approximately two years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/complex-multiple-long-term-conditions-mltc-phenotypes-trends-andendpoints-compute

Complex Multiple long-term conditions (MLTC) Phenotypes, Trends, and Endpoints (CoMPuTE)

Last updated:
ID:
108536
Start date:
27 February 2024
Project status:
Current
Principal investigator:
Professor Derrick Bennett
Lead institution:
University of Oxford, Great Britain

More than a quarter of adults in England have more than one health condition. By 2035 this is expected to increase by 10-17%. Having more than one condition is called ‘multiple long-term conditions’ (MLTC). The more conditions someone has, the more disabling the effects. MLTC is difficult for both patients and carers: taking more medicines (with possible problems caused by conflicting or simply too many medications); the cost and wasted time of attending too many healthcare appointments; and the day-to-day challenges of living with multiple conditions. This study hopes to predict who will suffer from MLTC, how MLTC will progress over a person’s lifetime and identify risk factors and their causal relevance for MLTC.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/complex-trait-analysis-from-genotype-and-low-pass-cell-free-and-ancient-dna-sequence-data-in-structured-populations

Complex trait analysis from genotype, and low-pass cell-free and ancient DNA sequence data in structured populations.

Last updated:
ID:
95216
Start date:
28 March 2023
Project status:
Current
Principal investigator:
Professor Toomas Kivisild
Lead institution:
Katholieke Universiteit Leuven, Belgium

Complex traits are influenced both by environmental and genetic factors. Typically, the genetic component of a complex trait consists of many genetic variants, which each have a small effect on the trait. A polygenic risk score combines the effect of these variants into a single genetic risk score, thus representing someone’s genetic risk to develop the disease or show a particular trait. As such, these scores are promising tools to be used in clinic and precision medicine. A polygenic risk score is most commonly calculated from high quality SNP array data. Low-coverage sequence data, such as maternal cell-free DNA from non-invasive prenatal testing (NIPT, typically performed to detect fetal abnormalities such as trisomy 21 in early pregnancy) or ancient DNA would however be an important alternative source. Since both the maternal cfDNA and ancient DNA are of very low concentration, missing genotypes need to be imputed. It is not clear, however, what impact sequence coverage and damage, fetal fraction and other factors affecting imputation accuracy altogether have on the PRS estimation of different phenotypic traits in the presence of different levels of population stratification. In this project we will use both genotype and low-pass sequence data from NIPT and ancient DNA samples of different genetic ancestries to assess the performance of different imputation and polygenic risk score estimation tools. Complex traits with different genetic architecture represented in the UK Biobank data will be used to evaluate the accuracy and potential biases in inference of polygenic risk scores. Our main clinically relevant focus will be on pregnancy related complex traits such as preeclampsia, SLE, gestational diabetes. In addition, other clinical phenotypes of interest, such as cancer (e.g. breast cancer), neurological diseases (e.g. Alzheimer’s disease), immune disorders (e.g. inflammatory bowel disease) will also be considered.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/complex-trait-prediction-from-multi-source-data-using-machine-learning-methods

Complex trait prediction from multi-source data using machine learning methods

Last updated:
ID:
81475
Start date:
22 February 2022
Project status:
Current
Principal investigator:
Dr Catherine Ruth Jutzeler
Lead institution:
ETH Zurich, Switzerland

Can we predict a participant’s height based on their genetics? Their environmental conditions? Or some combination of those things? Does their health status/history help or exacerbate these factors?

These are the questions we try to answer in our proposed three year project. Height is a so-called complex trait, which are traits that cannot be explained by simple genetic inheritance rules, and cannot be easily predicted. They are influenced by multiple factors such as genotypic data and environmental conditions. For example, a person’s height is influenced by the height of their parents (genotypic data), but also by other factors such as their diet (environmental conditions). Understanding a participants health status/history can further help explain a complex trait like height.

Each of these factors provides a different, incomplete view of a patient. However, the degree to which each of those factors, and combinations of these factors, affect a complex trait like height is still an open question in the scientific and medical community.

As health data becomes increasingly more comprehensive, machine learning can be used to investigate the relationship between these data types and complex traits, by identifying patterns that are predictive of a complex trait of interest in large amounts of data. Our goal is to develop and use a machine learning algorithm that can incorporate these different snapshots of a participant, and determine which phenotypes they influence most.

Identifying the factors driving a particular complex trait is relevant for public health because it can inform the design making and recommendations of practitioners in the medical field, and specifically indicate which levers should be pulled to change a health outcome. Identifying complex traits that are affected mostly by genetics could indicate that early screening may be critical so that treatment begins at onset; on the other hand, if particular environmental factors have the greatest effect on another phenotype, this indicates that public health preventive interventions could be impactful. The common thread underlying each of these scenarios is actively understanding how these different factors play a role in a participant’s health.

The UK Biobank is an ideal partner for us in this research, with its vast troves of genotypic, health status and environmental data. We will predict complex traits by extracting meaningful patterns from these data sources using machine learning. In doing so, we will unlock a critical component in our progress towards personalized medicine.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/components-of-heritability-in-a-uk-biobank-cohort

Components of heritability in a UK Biobank cohort

Last updated:
ID:
16549
Start date:
1 March 2016
Project status:
Current
Principal investigator:
Dr Alkes Price
Lead institution:
Harvard School of Public Health, United States of America

We will analyze heritability of several polygenic traits. We will use existing methods and methods under development for partitioning heritability by functional annotation (e.g. cell-type-specific enhancer regions, gene pathways, etc.) to learn about underlying trait biology. We will also examine how SNP heritability varies across LD and MAF categories. Finally, we will evaluate missing heritability using new methods to estimate heritability explained by haplotypes, narrow-sense heritability (using PSMC) and epistatic components of heritability (using Hadamard products). We plan to study a wide range of health-related phenotypes, including diseases and quantitative traits like height and BMI. The data in the UK Biobank?s cohort will allow us to partition heritability at higher resolution and to evaluate missing heritability. These will inform both our understanding of trait biology and the design of future genetic studies. Both of these outcomes will benefit attempts to find actionable drug targets for human disease. Moreover, the methods we develop for partitioning heritability will be published and made open-source for use by the broader research community. We will work with annotations from Finucane et al. 2015 Nat Gen as well as gene sets and new annotations from the ENCODE and Roadmap Epigenomics Consortia and others. We will apply LD score regression [Finucane et al. 2015 Nat Genet], BOLT-REML [Loh et al. 2015 Nat Genet], and a new method under development to assess heritability enrichment of these annotations, as well as enrichment/depletion by LD and MAF, within/across traits and populations. We will also apply new methods to estimate heritability explained by haplotypes, total narrow-sense heritability and epistatic components of heritability. We will analyze the full cohort.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/composite-polygenic-risk-score-analysis-of-trans-ethnic-portability-and-within-family-validation-of-prediction-using-compressed-sensing-and-high-dimensional-statistical-methods-in-complex-traits

Composite polygenic risk score analysis of trans-ethnic portability and within-family validation of prediction, using compressed sensing and high-dimensional statistical methods in complex traits.

Last updated:
ID:
54679
Start date:
19 October 2022
Project status:
Current
Principal investigator:
Mr Laurent Tellier
Lead institution:
Genomic Prediction, Inc., United States of America

Our goal is to develop and test new computational methods for determining the genetic contribution to complex traits, including highly heritable conditions such as Diabetes, Alzheimer’s Disease, Breast Cancer, Schizophrenia, and Major Depressive Disorder. Most traits are caused by both environmental components and a genetic component. The latter is usually the sum total of the contribution from many (up to tens of thousands of) genes.Through machine learning algorithms, we use DNA-information alone to predict traits. These disease risk predictions are not perfect, but can be of huge importance when integrated into clinical treatment and prevention. For example, genetic breast cancer prediction can inform women at extra high risk to begin early screening, and can thus alone save thousands of lives yearly.

Our research is both focused on producing the best possible predictors for the most important diseases, as well as for people of all ancestries, via further methodological development. We are also advancing the research on how to simultaneously combine multiple disease risk predictions into genomic indices, and how to best deploy such indices in a clinical application context.

There is already a real impact through the early clinical adaptations, and the coming health benefits for both individuals and the society at large are enormous. This project has a commitment to advance the field as long as there are scientific gains to be made for both a deeper understanding of the genetic nature and the best possible predictors of complex traits.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comprehensive-analysis-of-cardiovascular-disease-identifying-risk-factors-enhancing-diagnosis-optimizing-treatment-and-improving-prognosis

Comprehensive Analysis of Cardiovascular Disease: Identifying Risk Factors, Enhancing Diagnosis, Optimizing Treatment, and Improving Prognosis

Last updated:
ID:
216397
Start date:
3 January 2025
Project status:
Current
Principal investigator:
Dr Xiangwei Liu
Lead institution:
Xiangya Hospital of Central South University, China

Title: “Unlocking Insights into Cardiovascular Health: A Comprehensive Study Using UK Biobank Data”
Aims: Our project aims to delve into the complexities of cardiovascular diseases (CVD) – a leading cause of health issues worldwide. Using the UK Biobank’s vast dataset, we plan to explore four main areas: identifying risk factors, enhancing disease diagnosis, evaluating treatment effectiveness, and improving predictions about disease progression and patient outcomes.
Scientific Rationale: Cardiovascular diseases, like heart attacks and strokes, are major health concerns globally. Understanding them better is crucial for improving how we prevent, diagnose, and treat these conditions. Our project taps into the UK Biobank’s extensive data, including genetic information, lifestyle factors, and medical records, to gain deeper insights into these diseases.
project Duration: We anticipate that this study will span over a period of three years. This duration will allow us sufficient time to thoroughly analyze the data, interpret our findings, and share these insights with the medical community and the public.
Public Health Impact: The impact of our study on public health could be significant. Here’s how:
1. Better Prevention: By identifying key risk factors, we can inform public health campaigns and personal lifestyle choices, potentially preventing many cases of CVD.
2. Improved Diagnosis: Developing more accurate diagnostic tools means people with CVD can be identified earlier and treated more effectively, increasing their chances of a better health outcome.
3. Personalized Treatments: Understanding how different treatments work for various groups can lead to more personalized, effective care plans.
4. Accurate Predictions: Better predictions about how a patient’s disease will progress can help doctors and patients make more informed decisions about their care.
In simple terms, our project aims to use the wealth of data in the UK Biobank to understand heart and blood vessel diseases better. Our goal is to make a real difference in how these diseases are prevented, detected, and treated, ultimately improving the health and wellbeing of people not just in the UK, but around the world.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comprehensive-analysis-of-cardiovascular-diseases-cancers-musculoskeletal-disorders-and-neurodegenerative-diseases-using-health-genomic-and-imaging-data-from-the-uk-biobank

Comprehensive Analysis of Cardiovascular Diseases, Cancers, Musculoskeletal Disorders, and Neurodegenerative Diseases Using Health, Genomic, and Imaging Data from the UK Biobank

Last updated:
ID:
737152
Start date:
9 April 2025
Project status:
Current
Principal investigator:
Dr Jinmin Liu
Lead institution:
LanZhou University, China

This project aims to leverage the extensive data available from the UK Biobank to investigate the common mechanisms underlying cardiovascular diseases, cancers, musculoskeletal disorders, and neurodegenerative diseases. These diseases represent some of the leading causes of morbidity and mortality worldwide, yet the complex genetic, environmental, and clinical factors contributing to their pathogenesis remain poorly understood.
Our primary research question is: How do genetic, lifestyle, and imaging factors interact to influence the risk and progression of cardiovascular diseases, cancers, musculoskeletal disorders, and neurodegenerative diseases? Specifically, we will:
1. Identify genetic variants associated with these diseases through genome-wide association studies (GWAS).
2. Investigate the relationships between clinical data (e.g., medical history, lifestyle factors) and disease onset or progression.
3. Analyze the role of imaging data in the detection of structural changes related to these diseases, with an emphasis on neurodegenerative and cardiovascular conditions.
The scientific rationale behind this study is to bridge the gap in understanding the intersection of genetics, clinical factors, and imaging biomarkers in disease pathogenesis. By integrating these data, we aim to improve early detection strategies, identify novel biomarkers for disease prediction, and provide insights into personalized treatment approaches. This research is essential for advancing precision medicine, particularly in diseases that currently have high morbidity and mortality rates, such as cardiovascular diseases, cancers, and neurodegenerative conditions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comprehensive-analysis-of-determinants-in-non-communicable-and-communicable-diseases-exploring-genetic-environmental-clinical-laboratory-imaging-and-lifestyle-factors

Comprehensive Analysis of Determinants in Non-Communicable and Communicable Diseases: Exploring Genetic, Environmental, Clinical, Laboratory, Imaging, and Lifestyle Factors

Last updated:
ID:
663576
Start date:
18 March 2025
Project status:
Current
Principal investigator:
Dr Shenshen Huang
Lead institution:
Henan University of Science and Technology, China

This project aims to conduct a comprehensive analysis of key determinants influencing the onset and progression of both non-communicable diseases (NCDs) and communicable diseases (CDs). It will explore genetic, environmental, clinical, laboratory, imaging, and lifestyle factors and investigate their interactions in shaping disease outcomes.

Research Questions:

1. What are the key genetic, environmental, clinical, and lifestyle factors contributing to the onset and progression of NCDs (e.g., cardiovascular diseases, diabetes) and CDs (e.g., infectious diseases, sepsis)?
2. How do these determinants interact, and what are their combined effects on disease risk and progression?
3. Can identifying these factors provide novel insights for personalized prevention and therapeutic strategies?

Objectives:

1. Identify and analyze genetic, environmental, clinical, laboratory, imaging, and lifestyle determinants for NCDs and CDs.
2. Investigate the interactions between these factors and their combined impact on disease risk and progression.
3. Provide insights for personalized prevention, early diagnosis, and therapeutic interventions.
Scientific Rationale: The increasing global burden of NCDs and CDs necessitates a deeper understanding of their multifactorial nature. By examining the interplay between genetic predispositions, environmental exposures, lifestyle factors, and clinical data, this research aims to uncover the mechanisms underlying disease development. This will contribute to precision medicine by offering tailored interventions and improving public health outcomes. Furthermore, the findings may help identify new drug targets and diagnostic tools for these diseases, advancing our understanding of their pathogenesis.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comprehensive-analysis-of-genetic-metabolic-proteomic-and-environmental-determinants-in-cancer-risk-and-prognosis

Comprehensive Analysis of Genetic, Metabolic, Proteomic, and Environmental Determinants in Cancer Risk and Prognosis

Last updated:
ID:
709086
Start date:
30 July 2025
Project status:
Current
Principal investigator:
Dr Dennis Hsu
Lead institution:
University of Pittsburgh, United States of America

Cancer pathogenesis and outcome are influenced by a complex interplay of genetic and environmental factors. Understanding these factors is of paramount importance in identifying at-risk individuals and developing effective preventive and therapeutic approaches. Through adopting an omics approach, this project aims to address how the genetic and environmental factors individually and interactively affect cancer pathogenesis, prognosis, and response to treatment. Through leveraging advanced statistical methods and machine learning, we will evaluate the effects of genetic variants, environmental risk factors, and metabolic and proteomics variables on the development and progression of cancer based on the UK Biobank Database. In addition, we aim to investigate the interactive effect of these factors in the final causal network.
This project has important public health implications by identifying novel risk factors in cancer predisposition and progression and offering preventive and treatment avenues to decrease the incidence of cancer and improve the survival rate of those with cancer, ultimately reducing the cancer burden on society. Our findings can also pave the way for the advancement of precision medicine in cancer care and prevention.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comprehensive-analysis-of-laboratory-risk-factors-and-genetic-associations-for-postherpetic-neuralgia-focus-on-inflammatory-blood-biomarkers

Comprehensive Analysis of Laboratory Risk Factors and Genetic Associations for Postherpetic Neuralgia: Focus on Inflammatory Blood Biomarkers

Last updated:
ID:
956257
Start date:
4 November 2025
Project status:
Current
Principal investigator:
Miss Yike Liu
Lead institution:
Southern Medical University, China

Postherpetic neuralgia (PHN), as the most challenging complication of herpes zoster, is closely associated with intense and persistent inflammatory responses triggered by viral reactivation. Scientific evidence clearly demonstrates that the reactivation of varicella-zoster virus (VZV) in sensory ganglia not only causes skin lesions but, more critically, initiates a severe inflammatory storm that spreads to the peripheral and even central nervous systems. The key players in this inflammation-pro-inflammatory cytokines and immune cells-exhibit detectable changes in their activity and concentration in the blood, providing a crucial window for identifying predictive and diagnostic laboratory markers for PHN. Studies reveal that PHN patients commonly exhibit significant inflammatory imbalances in their blood: levels of key pro-inflammatory cytokines such as interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-A), interleukin-1 beta (IL-1B), and interleukin-17 (IL-17) are markedly and persistently elevated. These cytokines not only directly act on damaged nerves, increasing their excitability and inducing ectopic discharges, but also drive peripheral and central sensitization, amplifying pain signals. Concurrently, impaired immune regulation is evidenced by reduced numbers or functionality of regulatory T cells (Tregs) in the blood, while pro-inflammatory T helper 17 cells (Th17) are relatively increased, leading to an imbalanced Th17/Treg ratio that further exacerbates the inflammatory environment. Additionally, nonspecific systemic inflammatory markers such as C-reactive protein (CRP) and erythrocyte sedimentation rate (ESR) are often elevated during the acute phase or in patients with persistent pain and correlate with pain intensity. Beyond cytokines and immune cell profiles, pain-related neuropeptides like substance P (SP) and calcitonin gene-related peptide (CGRP) are also abnormally elevated in the serum of PHN patients, directly participating in the transmission


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comprehensive-analysis-of-lifestyle-genetic-and-environmental-factors-on-urological-tumors-integrating-clinical-data-imaging-studies-and-genetic-analyses-using-the-uk-biobank-cohort

Comprehensive Analysis of Lifestyle, Genetic, and Environmental Factors on Urological Tumors: Integrating Clinical Data, Imaging Studies, and Genetic Analyses Using the UK Biobank Cohort

Last updated:
ID:
322987
Start date:
12 November 2024
Project status:
Current
Principal investigator:
Dr Changning Lv
Lead institution:
Peking University First Hospital, China

Project Summary:
This study aims to analyze the extensive data from the UK Biobank to identify how lifestyle, genetic, and environmental factors affect the occurrence and development of urological tumors (such as bladder cancer and kidney cancer). We will comprehensively use clinical data, genetic data, and imaging data to deeply understand the mechanisms through which these factors influence tumors.

Project Objectives:
Our goal is to identify which lifestyle habits (such as diet, exercise, smoking), genetic characteristics, and environmental exposures (such as chemical exposure) increase the risk of urological tumors. Through this research, we hope to provide scientific evidence to help prevent, detect early, and personalize the treatment of these tumors.

Scientific Principles:
This project is based on the fundamental principles of epidemiology, genetics, and oncology. We will use advanced statistical methods and bioinformatics tools to analyze large amounts of data to identify key factors and mechanisms that may lead to tumors. These analyses will help us understand which genes and lifestyle habits have the greatest impact on tumors.

Project Duration:
The project is expected to last for three years. In the first year, we will collect and organize the data. In the second year, we will conduct data analysis and model construction. In the third year, we will validate the research results and write the report.

Public Health Impact:
By identifying key risk factors for urological tumors, our research will help develop more effective prevention measures and reduce the incidence of tumors. Additionally, the results will aid in early screening and personalized treatment, improving patient survival rates and quality of life. Ultimately, this will help improve public health policies and promote overall societal health.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comprehensive-analysis-of-treatment-patterns-and-prognosis-prediction-in-breast-cancer-patients-using-the-uk-biobank-database

Comprehensive Analysis of Treatment Patterns and Prognosis Prediction in Breast Cancer Patients Using the UK Biobank Database

Last updated:
ID:
586650
Start date:
20 April 2025
Project status:
Current
Principal investigator:
Dr Qingyao Shang
Lead institution:
Chinese Academy of Medical Sciences &Peking Union Medical College, China

Breast cancer is a heterogeneous disease with diverse molecular subtypes, gene expression profiles, and immune infiltration, influencing treatment responses. Currently, numerous breast cancer treatment options have been developed, including chemotherapy, endocrine therapies, CDK4/6 inhibitors, PI3K inhibitors, HER2-targeted agents, ADCs, and immunotherapies. The complexity of disease and treatment underscores the need for personalized strategies to improve recurrence and survival outcomes.
1.Research Questions:
What clinical and molecular factors influence treatment response across drug classes?
How do specific therapies and combinations affect recurrence and survival in breast cancer subtypes?
Can a dynamic model predict recurrence using time-dependent clinical, genetic, and immune factors?
What is the efficacy of combination therapies compared to monotherapy in improving survival?
How can a comprehensive mortality risk model guide personalized treatment?
2.Objectives:
Identify key clinical and molecular markers affecting treatment response.
Analyze drug choices and survival outcomes, including targeted therapies.
Develop a dynamic model for predicting recurrence patterns.
Assess combination therapies’ effectiveness in enhancing survival.
Build a comprehensive risk model for breast cancer-specific mortality.
3.Scientific Rationale:
This study leverages UK Biobank’s clinical and molecular data to address the complexity of breast cancer treatment. Identifying factors influencing outcomes will optimize drug selection, refine personalized strategies, and improve long-term survival. The findings aim to inform therapeutic guidelines, reduce recurrence, and enhance patients’ quality of life.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comprehensive-evaluation-of-causal-relationship-among-different-phenotypes-and-disease-traits-by-mendelian-randomization-using-genetic-instruments

Comprehensive evaluation of causal relationship among different phenotypes and disease traits by Mendelian Randomization using Genetic Instruments

Last updated:
ID:
66813
Start date:
28 April 2021
Project status:
Current
Principal investigator:
Dr Yung Na
Lead institution:
University of Hong Kong, Hong Kong

Most of the previous studies focused on the association between several risk factors and disease traits. Few studies demonstrated the cause-and-effect relationship, especially in a larger population. Our previous studies based on large-scale datasets suggested significant associations among different types of diseases and cancers. This suggests that an individual who has one type of disease/cancer (primary disease) may be at increased risk for another type of diseases/cancer (secondary disease). Such associations may be caused by potential biological mechanisms common in both diseases and shared environmental factors such as smoking. Therefore, in the current study, we aim to assess the causal relationship between exposures (such as disease traits etc.) and other diseases adjusting for potential shared factors (such as smoke, alcohol, blood glucose, etc.) by using a new statistical approach: Mendelian Randomization (MR). Such an approach allows us to interpret the causal relationship between two components: exposures and outcomes.
We will spend 36 months to perform this comprehensive analysis.
Results from our study will have several public health impacts: 1) the causal relationship between one disease and another disease may help us apply early protection/screening in patients with the disease; 2) the causal relationship may provide important evidence of biological mechanisms worth further investigation via a series of experiments, and may help us target important genetic variants associated with the diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comprehensive-evaluation-of-direct-contributions-of-rare-and-common-variants-to-genetic-risk-prediction

Comprehensive evaluation of direct contributions of rare and common variants to genetic risk prediction

Last updated:
ID:
103244
Start date:
9 June 2023
Project status:
Current
Principal investigator:
Mr Spencer Michael Moore
Lead institution:
Heliospect Genomics LLC, United States of America

Complex traits and diseases, like Type II diabetes, coronary artery disease, and schizophrenia, come from a mix of environmental and genetic factors. Until recently, it has been difficult to understand these factors well enough to predict these diseases using genetic information. We want to improve genetic risk predictions for these diseases and others by using advanced techniques on new genetic data, including whole-exome and whole-genome data. We will also consider how environmental factors interact with genetics and how they might be related.
To do this, we’ll compare the accuracy of genetic risk scores based on family and population data. If the scores from family data are less accurate, it means environmental factors related to genetics might be affecting the results. These factors can come from population differences or how parents’ genes create different environments for their children. By using family data, we can avoid these issues.
On the other hand, gene-environment interactions suggest that the real effects of genes might change depending on other factors, like socioeconomic status. This is because socioeconomic status can affect how genes contribute to diseases or traits. To improve risk predictions, we need to consider these factors and adjust the importance of genetic risks accordingly.
However, our understanding of these interactions is still new, and we don’t know if our findings will apply to rare genetic variants or different populations. To address these questions, we will use a variety of approaches, including family-level analysis, on the latest available data. This will help us answer broader questions about how genes and environments are connected.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comprehensive-evaluation-of-the-interaction-between-life-course-risk-factors-and-multi-omics-on-common-human-diseases

Comprehensive evaluation of the interaction between life course risk factors and multi-omics on common human diseases

Last updated:
ID:
188093
Start date:
13 February 2025
Project status:
Current
Principal investigator:
Dr Xiang Zhou
Lead institution:
The Second Affiliated Hospital of Nanjing Medical University., China

The prevalence of common diseases like cardio-cerebrovascular diseases, respiratory diseases, metabolic diseases, and certain cancers continues to rise globally, posing significant challenges to public health. Research indicates that these diseases are influenced not only by genetic factors but also by environmental and lifestyle factors across an individual’s life course. Understanding the intricate interplay between life course risk factors and multi-omics (genomics and metabolomics, etc.) could provide crucial insights into disease etiology, progression, and potential interventions. This study aims to identify key life course risk factors contributing to the onset and progression of common diseases, investigate how multi-omics data (e.g., genomics and metabolomics, etc.) interact with and are influenced by these risk factors, and analyze the potential synergistic or antagonistic effects of these interactions on disease susceptibility and severity. The project duration is 36 months. This research may lead to improved disease management and public health interventions, potentially reducing the burden of common diseases on a global scale.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comprehensive-gene-environment-interaction-analysis-in-chronic-diseases-and-cancers

Comprehensive gene-environment interaction analysis in chronic diseases and cancers

Last updated:
ID:
100359
Start date:
20 June 2023
Project status:
Current
Principal investigator:
Dr Kyeongmin Kwak
Lead institution:
Korea University College of Medicine, Korea (South)

Extensive research within the last several decades has revealed that the major risk factors for most chronic diseases including cancers are infections, obesity, alcohol, tobacco, radiation, environmental pollutants, and diet. It is now well established that these factors induce chronic diseases through induction of inflammation. Moreover, Inflammation is often associated with the development and progression of cancer. The chronically inflammation can activate signaling pathways that cross-talk between inflammation and carcinogenesis. Therefore, environmental factors are increasingly being studied for their associations with chronic diseases and cancers. Genetic factors also underlie the differential vulnerability to environmental risk factors of susceptible individuals. Currently the way in which environmental risk factors interact with genetic factors to increase the incidence of chronic diseases and cancers is not well understood. In this study, we aim to conduct a wide range of interaction studies on genetic variations and environmental factors including lifestyle and occupations related to developing inflammatory diseases and cancers. In addition, by restricting various operational definitions, researchers intend to conduct gene-environment interactions for chronic diseases including cancers and environmental factors. Especially, for air pollutants, which are major environmental exposure, we plan to perform gene-environment interaction analysis for chronic disease and cancer susceptibility and genetic variants by linking air pollutants measurement data in the UK. We plan to conduct these comprehensive analyses by running this project for 3 years (2023-2026). Findings regarding gene-environment interactions have a significant public health implication in disease prevention, as they can help identify groups of individuals who are more likely to benefit from lifestyle or environmental modifications.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comprehensive-genetic-correlation-of-disease-from-the-point-of-germline-structural-variations-associated-with-clinical-phenotypes

Comprehensive genetic correlation of disease from the point of germline structural variations associated with clinical phenotypes

Last updated:
ID:
98447
Start date:
17 April 2023
Project status:
Current
Principal investigator:
Professor Soyeon Ahn
Lead institution:
Seoul National University Bundang Hospital, Korea (South)

The aim of the study is to uncover novel associations between common germline structural variations (SVs) and various traits and diseases and to replicate the associations between germline structural variations and clinical traits found in 2,500 Koreans. Through the study, we will be able to understand the nature of germline SVs associated with clinical traits such as the genomic regions that are frequently subject to trait-associated structural variations and what their effect sizes will be, etc. The trait or disease associated SVs will be incorporated into polygenic risk score that usually contain single nucleotide variations (SNVs) and will enable more accurate and reliable prediction of diseases for early detection and prevention of diseases. The study will last for about three years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comprehensive-genome-exposome-phenome-association-studies-for-complex-diseases

Comprehensive genome-exposome-phenome association studies for complex diseases

Last updated:
ID:
37072
Start date:
3 September 2018
Project status:
Closed
Principal investigator:
Professor Isaac Kohane
Lead institution:
Harvard School of Public Health, United States of America

A comprehensive study on the risk factors of common diseases, including hypertension, diabetes, heart diseases, stroke, cancers, and psychiatric disorders, is lacking. In this study, we will identify the correlations between the participants’ genetic backgrounds / environmental exposures and the risk of developing common diseases. We will also associate genetic and environmental factors with participants’ blood and urine assay, and medical imaging results (such as MRI and bone density), which is expected to quantify the molecular and anatomical impact of the exposures. We expect to complete the proposed project in 36 months.

Our project aims at identifying the associations between genotypes, environmental factors, and complex diseases systematically. Our objectives are aligned with UK Biobank’s goal to improve the prevention, diagnosis, and treatment of a wide range of serious and life-threatening illnesses.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comprehensive-genome-wide-association-study-of-different-forms-of-hernia

Comprehensive genome-wide association study of different forms of hernia

Last updated:
ID:
33395
Start date:
22 February 2018
Project status:
Closed
Principal investigator:
Mr Frank Geller
Lead institution:
Statens Serum Institut, Denmark

The term hernia summarizes connective tissue ruptures that result in tissue leaving its normal position by passing through the opening. Hernias are divided by their location. The condition might not cause any symptoms but the opening often grows over time and can become a problem. More than 50,000 records of hernia can be identified from ICD diagnoses and operation codes (diaphragmatic: >24,000, inguinal: >18,000, umbilical: > 4,700, ventral: >2900, femoral: >600). The resulting case numbers will empower genetic studies of all and individual hernias. The project investigates the genetics of several more or less common forms of hernia. By now, only a genome-wide association study of inguinal hernia has identified robustly associated genetic variants. Standard genome-wide association studies for the different forms of hernia and the combined group of any hernia will be performed, comparing allele frequencies in individuals who fit the case definition vs. the remaining individuals as controls. Initially, the full cohort is investigated. The age structure of the cases might lead to certain restrictions in terms of age and sex for the controls, as younger unaffected individuals could be much more likely to develop the condition later in life than older ones.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comprehensive-genomic-and-proteomic-profiling-of-pancreatic-cancer-insights-from-uk-biobank-data-with-clinical-validation

Comprehensive Genomic and Proteomic Profiling of Pancreatic Cancer: Insights from UK Biobank Data with Clinical Validation

Last updated:
ID:
687634
Start date:
17 April 2025
Project status:
Current
Principal investigator:
Dr Sen Yang
Lead institution:
Peking Union Medical College Hospital (PUMCH), China

Research Questions:
1. What are the key genomic and proteomic alterations associated with the development and progression of pancreatic cancer?
2. How do these molecular features correlate with demographic, environmental, and lifestyle factors in the UK Biobank dataset?
3. Can findings from the UK Biobank data be used to identify potential biomarkers for clinical application?

Objectives:
1. To analyze the genomic and proteomic data of pancreatic cancer cases in the UK Biobank to identify common mutations (e.g., KRAS, TP53, CDKN2A, SMAD4) and protein expression changes.
2. To assess associations between molecular alterations and participant characteristics such as age, sex, environmental exposures, and lifestyle factors.
3. To generate hypotheses and insights that will be validated using independent clinical samples outside the UK Biobank.

Scientific Rationale:
Pancreatic cancer is characterized by a poor prognosis and limited effective biomarkers for early detection or targeted therapy. Using the comprehensive genomic and proteomic data within the UK Biobank, this project aims to uncover critical molecular alterations associated with pancreatic cancer. These findings will contribute to a better understanding of disease mechanisms and provide a foundation for the development of novel biomarkers or therapeutic strategies. Importantly, the UK Biobank data will be used exclusively for hypothesis generation, and clinical validation will be performed independently, ensuring non-depletable use of UK Biobank resources.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comprehensive-identification-of-risk-factors-and-biomarkers-for-musculoskeletal-disorders-osteoporosis-osteoarthritis-and-rheumatoid-arthritis

Comprehensive identification of risk factors and biomarkers for musculoskeletal disorders (osteoporosis, osteoarthritis, and rheumatoid arthritis)

Last updated:
ID:
108866
Start date:
21 March 2024
Project status:
Current
Principal investigator:
Professor Dezhi Tang
Lead institution:
Shanghai University of Traditional Chinese Medicine, China

Stating the aims:
This project aims to (1) investigate the relationship between genetics, behavior, diet structure, lifestyle, cognition, environmental factors, biomarkers and musculoskeletal diseases (osteoporosis, osteoarthritis, rheumatoid arthritis, etc.); (2) further identify potential pathways between various exposures and musculoskeletal diseases such as osteoporosis, osteoarthritis, and rheumatoid arthritis; (3) use genetic information, multi-omics sequencing, and anthropometry to predict the occurrence and progression of musculoskeletal-related diseases; (4) explore the internal biological mechanism of the interaction between common chronic diseases (hypertension, diabetes, psychiatric disorders, etc.) and musculoskeletal diseases.
Method: Strict statistical analysis methods (Cox proportional hazard model, Logistics regression model, machine learning algorithm, etc.) will be used to identify valuable genetic, lifestyle and other factors. According to the results of multi-group sequencing, the potential targets and biomarkers of musculoskeletal diseases were explored, the targeted intervention measures were clarified, and the potential mechanism was further clarified.
Project duration: The duration of this project is approximately 3 years.
Scientific rationale: It is well known that musculoskeletal disorders are influenced by a combination of genetic factors, lifestyle, environmental factors and chronic co-morbidities. However, exposure affects different groups differently, depending on factors such as age, sex, or genetics. Based on genetic and acquired factors, early identification of high-risk individuals and accurate prevention and treatment of bone diseases are of great significance.
Public health impact: A comprehensive understanding of risk factors for skeletal diseases will help identify high-risk groups, intervene early, effectively reduce the harm of skeletal diseases, reduce social and personal costs, and improve the quality of life of individuals.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comprehensive-impact-of-social-factors-lifestyle-genetics-environmental-exposures-and-plasma-biomarkers-on-the-incidence-and-prognosis-of-common-non-communicable-chronic-diseases

Comprehensive impact of social factors, lifestyle, genetics, environmental exposures, and plasma biomarkers on the incidence and prognosis of common non-communicable chronic diseases

Last updated:
ID:
475786
Start date:
11 November 2024
Project status:
Current
Principal investigator:
Dr Jie Bai
Lead institution:
Chongqing Medical University, China

The proposed research aims to investigate the integrated impact of social factors, lifestyle choices, genetic predispositions, environmental exposures, and plasma biomarkers on both the incidence and prognosis of common non-communicable chronic diseases (NCD), including cardiovascular disease, diabetes, and cancer. This study will utilize UK Biobank’s extensive data to achieve the following objectives:
1. Data Integration and Analysis: We will conduct a comprehensive analysis of UK Biobank’s data to explore how social determinants (such as socioeconomic status and education), lifestyle factors (including diet, physical activity, and smoking), genetic markers, environmental exposures (such as air pollution and occupational hazards), and plasma biomarkers interact to influence the risk of developing NCD. Advanced statistical models and machine learning techniques will be employed to identify significant associations and interactions.
2. Assessment of Disease Progression: Using longitudinal data, we will assess how these factors contribute to the progression and prognosis of NCD. This will involve evaluating the impact of early-life exposures and current risk factors on disease outcomes, including disease severity, complications, and mortality rates.
3. Identification of Predictive Biomarkers and Risk Profiles: We will focus on identifying biomarkers and risk profiles that can predict disease onset and progression. This will include analyzing plasma biomarker data to uncover potential diagnostic and prognostic markers. The study aims to develop risk prediction models that integrate genetic, environmental, and biological factors to inform personalized prevention and treatment strategies.
The findings from this research will provide valuable insights into the multifaceted causes of NCD and could lead to more effective public health interventions and personalized medical approaches.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comprehensive-investigation-in-common-disease-outcomes-integrating-genetic-and-phenotypic-associations

Comprehensive investigation in common disease outcomes integrating genetic and phenotypic associations

Last updated:
ID:
73759
Start date:
9 August 2021
Project status:
Current
Principal investigator:
Dr Yazhou He
Lead institution:
Sichuan University, China

Most common diseases, such as cardiovascular diseases and cancer, are conditions attributed to intricated interplays between human DNA and environmental determinants. The UK Biobank has provided an immense resource of multidimensional data (e.g. human DNA variations and environmental factors) enabling identification of novel predictors for common diseases with considerable clinical and public health relevance. This also permits systematic combined assessment of the human DNA, environmental factors, imaging data and other markers for disease risk and clinical outcome after diagnosis. In this project, firstly we aim to unravel novel markers that lie in human DNA as well as environmental factors associated with common disease risk and outcomes using cutting-edge statistical methods. Novel findings along with previously reported marker-disease associations will then be further appraised for the strength of evidence and be evaluated for possible causality. The third objective is to improve prediction accuracy of disease risk and outcomes integrating all the identified markers using modern data science techniques such as machine learning. A special focus will be put on common diseases that impose substantial burdens on human health such as cardiovascular diseases, cancer, infections, autoimmune and aging related diseases. Combination of the big data provided by the UK Biobank and newly-developed modelling methodology will add to current knowledge of pathogenesis and progression of common diseases. Our findings may also provide evidence for individualised disease prevention and management. This project may be extended beyond three years due to newly-released data which warrant extra validation of our findings.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comprehensive-investigation-into-the-relationship-of-genetic-and-environmental-factors-with-metabolic-diseases-and-cancer

Comprehensive investigation into the relationship of genetic and environmental factors with metabolic diseases and cancer

Last updated:
ID:
78619
Start date:
24 February 2022
Project status:
Current
Principal investigator:
Dr Liyuan Han
Lead institution:
Ningbo University, China

Aims: Our research aims to use raw data from UK Biobank to systematically investigate the complex relationships of lifestyles, physical and biological measures, biomarkers and genetic susceptibility with metabolic diseases (e.g. hyperuricemia, diabetes, kidney disease) and cancer by using traditional statistical methods and machine learning algorithms.

Scientific rationale: The risk of metabolic diseases and cancer is determined by both environmental and genetic factors. However, there is still a paucity of data regarding the effects of gene-environment interaction on these diseases. Moreover, the mechanism of this risk remains unclear. In addition, traditional statistical methods often have inherent limitations in modeling the complex relationships between various risk factors and the clinical outcomes, whereas the advantages of machine learning techniques may help fill in this gap.

Project duration: This project will last for 60 months.

Public health impact: Findings from this research may provide significant healthcare benefits of adhering to a healthy lifestyle in individuals with metabolic diseases or cancer. Better understanding of the main risk factors of these diseases will enable physicians to identify high risk patients for early intensification and individualization of treatment to prevent these diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comprehensive-investigation-of-biomarkers-and-medical-conditions-in-relation-to-subsequent-hematological-disorders-or-cancer

Comprehensive investigation of biomarkers and medical conditions in relation to subsequent hematological disorders or cancer

Last updated:
ID:
106912
Start date:
20 July 2023
Project status:
Current
Principal investigator:
Professor Qianwei Liu
Lead institution:
Nanfang Hospital, Southern Medical University, China

The nature of disease is a continuous process but not a categorical definition. Investigating biomarker changes or medical conditions (or disease) in early stage before clinical onset of subsequent disease is of much importance, which could provide evidence for early intervention or screening and provide clue for understanding potential biological mechanism. In this project, we will mainly focus on diseases that are strongly related to serum biomarker changes including hematological disorders and cancer. To be more specific, we aim to: 1) systematically investigate influence factors (risk factors or protective factors) of the association between biomarkers or medical conditions (or disease) in earlier stage and subsequent hematological disorders and cancer in later stage, which could help to provide knowledge for disease prevention and screening; 2) identify novel biomarkers or early medical conditions that are related to subsequent hematological disorders and cancer, and explore the associations of biomarkers or medical conditions (or disease) that are related to hematological disorders and cancer with other disorders (beyond hematological disorders and cancer); 3) investigate impact of these biomarkers or medical conditions (or disease) in earlier stage on the prognosis of subsequent hematological disorders or cancer. Taken together, our findings could provide evidence for early intervention or screening and would be helpful for understanding biological basis for the associations of interest.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comprehensive-investigation-of-determinants-and-risk-factors-associated-with-the-development-and-progression-of-chronic-kidney-disease

Comprehensive investigation of determinants and risk factors associated with the development and progression of chronic kidney disease

Last updated:
ID:
88543
Start date:
9 August 2022
Project status:
Current
Principal investigator:
Dr Anand Srivastava
Lead institution:
University of Illinois at Chicago, United States of America

Chronic kidney disease (CKD) increases the risks of cardiovascular disease, end-stage kidney disease, and death. Emerging literature suggests that CKD worsens outcomes in patients with other comorbid conditions, such as lung disease, metabolic syndrome, liver disease, and cancer. Enhanced understanding of the relationships of CKD with other comorbid conditions will enable us to study factors associated with adverse clinical outcomes to eventually personalize therapy. We propose to perform a comprehensive analysis to evaluate lifestyle, social, environmental, genetic, biochemical, and imaging markers associated with the development of CKD and cardiovascular disease, progression of CKD, and death. We will also evaluate these outcomes in patients with pre-existing cardiovascular disease, lung disease, liver disease, metabolic syndrome, and cancer. We will also use all available data to identify subgroups of individuals with and without CKD to determine the risks for development of CKD and worsening CKD. The results of this study will identify novel determinants and factors associated with the development of CKD and worsening CKD in multiple disease states that will inform the design and conduct of clinical trials to personalize therapies to reduce the risk of adverse clinical outcomes.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comprehensive-investigation-of-lipidomics-and-its-genetic-determinants-in-cancer-risk-and-prognosis-by-evidence-triangulation

Comprehensive investigation of lipidomics and its genetic determinants in cancer risk and prognosis by evidence triangulation

Last updated:
ID:
87201
Start date:
25 August 2022
Project status:
Current
Principal investigator:
Dr Rayjean Hung
Lead institution:
Sinai Health System., Canada

The proposed study aims to understand the role of blood lipids in cancer development and progression. Blood lipids are fat-related molecules that can be measured in blood, such as cholesterol. They have long been associated with cancers, but results from previous studies have been inconsistent. With emerging technologies, lipidomics is a new field that can provide a complete analysis of lipids in the body. Therefore, we propose to assess the associations between a wide range of blood lipids and cancer outcomes using multiple approaches to strengthen the findings.

The project will take three steps. First, we will investigate the associations between lipids and cancer occurrence and progression. Next, we will identify genetic factors that are strongly associated with lipids in the UK Biobank. These genetic factors will then be used to evaluate the causal relationship between lipids and cancers in independent datasets. Third, we will develop a risk prediction model including the predictors related to lipids identified previously.

The proposed project will take about 2 years to complete. We anticipate 3-4 months to clean the data and set up the study cohort, 10-12 months to conduct the analyses, and 5-6 months to write the manuscripts and present at conferences.

This research project will have several public health impacts: 1) it will improve the understanding of blood lipids in cancers; 2) it will generate a risk prediction model that can be jointly used with clinical evaluations to better predict future cancer events; 3) the findings of the study can be used to design possible intervention strategies to reduce cancer risk and improve prognosis.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comprehensive-multi-omics-analysis-of-the-neuropsychiatric-system-to-uncover-mechanisms-and-therapeutic-targets

Comprehensive Multi-Omics Analysis of the Neuropsychiatric System to Uncover Mechanisms and Therapeutic Targets

Last updated:
ID:
858903
Start date:
24 June 2025
Project status:
Current
Principal investigator:
Miss Rong Wang
Lead institution:
Third Xiangya Hospital of Central South University, China

Neurological diseases, such as cognitive impairment and stroke, represent a growing global health concern, especially in aging populations. These disorders significantly reduce quality of life, contribute heavily to disability-adjusted life years (DALYs), and place an immense burden on healthcare systems worldwide. In addition to primary neurological conditions, comorbid psychiatric symptoms-such as depression and anxiety-frequently coexist, further complicating disease progression and management.
Recent advancements in high-throughput sequencing technologies and systems biology approaches have enabled multi-omics investigations into complex diseases. By integrating genomics, transcriptomics, epigenomics, metabolomics, and proteomics data, researchers can identify molecular networks and pathways that contribute to neuropsychiatric disorders.
The objective of this project is to conduct a multi-omics analysis of the neuropsychiatric system to identify biomarkers, biological pathways, and therapeutic targets associated with various mental health conditions. Key research questions include: 1. What molecular signatures are consistently associated with major neuropsychiatric disorders? 2. How do genomic and epigenomic variations influence transcriptomic and metabolic changes in the brain? 3. Can we identify novel therapeutic targets or predictive biomarkers using systems biology approaches?
This integrative research will leverage multi-omics datasets to build predictive models of disease, investigate potential causal pathways, and propose mechanisms for comorbidity among mental health disorders. The study aims to improve understanding of disease etiology, enhance early diagnostic accuracy, and facilitate the development of targeted therapeutic interventions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comprehensive-multi-omics-and-population-based-research-on-the-mechanistic-basis-and-precision-health-strategies-of-chronic-diseases

Comprehensive Multi-Omics and Population-Based Research on the Mechanistic Basis and Precision Health Strategies of Chronic Diseases

Last updated:
ID:
904444
Start date:
29 July 2025
Project status:
Current
Principal investigator:
Dr Weihua Li
Lead institution:
Peking University Health Science Center, China

Question: Chronic diseases are formally known as chronic non-communicable diseases. They do not specifically refer to any particular disease, but rather are a general term for a group of diseases that have insidious onset, long course, and persistent and unremitting conditions. Chronic non-communicable diseases, including cardiovascular, metabolic, neurodegenerative, respiratory, musculoskeletal, and oncological disorders! represent a major and growing global health burden. These conditions typically manifest with slow onset, prolonged progression, and complex, multifactorial etiologies that remain only partially understood. The lack of clearly defined infectious causes, coupled with the nonlinear interplay between genetic susceptibility, environmental exposures, and lifestyle factors, poses significant challenges to early detection and precision intervention.
Chronic diseases lack clear evidence of infectious biological causes, have complex etiologies, and some of their causes have not yet been fully confirmed. The main reasons why the pathogenesis is still not fully understood to this day are: 1. Multiple genetic and environmental factors are nonlinearly intertwined to form a complex pathological network; 2. Compensation mechanisms mask the early lesion characteristics; 3. There are unknown “critical qualitative change points” in the dynamic regulation of the life system. This leads to fundamental challenges in precise prevention and treatment.
Core Objective:
1. To leverage the full spectrum of UK Biobank resources-including genetic, phenotypic, environmental, imaging, biomarker, longitudinal health data, exposure factors and etc. to systematically elucidate the multifactorial mechanisms underlying chronic diseases and to develop personalized prevention and treatment strategies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comprehensive-phenotype-wide-association-studies-phewasfor-genetic-tool-variants-relevant-to-gsk-drug-targets

Comprehensive Phenotype-wide Association Studies (PheWAS)for Genetic Tool Variants relevant to GSK Drug Targets

Last updated:
ID:
20361
Start date:
13 June 2016
Project status:
Current
Principal investigator:
Dr Jonathan Davitte
Lead institution:
GlaxoSmithKline, USA, United States of America

Overall success rates for bringing novel medicines to patients are low. Reasons for failure in drug discovery and clinical development are many and complex, including choosing wrong target-indication pair(s) and limited understanding of the biology and mechanisms of action. It is now widely accepted that genetic associations with disease phenotypes may constitute ?drug target validation? evidence, with improved likelihood of success. This study aims to perform systematic Phenome-wide association studies (PheWAS) to evaluate associations between relevant drug-target genes and all health-related outcomes. All genotyping, health history, biochemistry and linked health-related outcomes will be requested. This research will systematically evaluate associations between relevant drug target gene variants and all health-related outcomes. This research will provide useful results to validate existing target-indication pairs, and discover alternative indications for existing drugs. Adding human target evidence in portfolio progression decisions may increase success rates in subsequent clinical development. Focusing investment in drugs/targets most likely to ultimately deliver patient benefit reduces patient numbers enrolled in trials that will ultimately fail (wasting patient volunteer effort and associated risks to patients). This fulfills the UK Biobank?s stated purpose to improve the prevention, diagnosis and treatment of illnesses. Variants and combinations of variants in existing and potential drug target genes will be evaluated for association with all health-related and disease outcomes as well as markers of disease severity and progression recorded within the UK Biobank. Initially, analyses will be performed using simple groupings of ICD-10 codes, followed by more detailed evaluations of disease outcomes and necessary sub-phenotypes. Genetic association analyses will utilize regression approaches, adjusting for potential confounders. Results will be submitted for publication in peer reviewed journals. We will request data for the full cohort


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comprehensive-risk-assessment-of-chronic-disease-onset-and-outcome

Comprehensive risk assessment of chronic disease onset and outcome

Last updated:
ID:
97846
Start date:
16 March 2023
Project status:
Current
Principal investigator:
Dr Mattias Johansson
Lead institution:
International Agency for Research on Cancer, France

Scientific rationale:
One in five people will develop a cancer during their lifetime. Cancer prevention has become one of the most important public health challenges of the 21st century. Based on current scientific evidence, at least 40% of all cancer cases can be prevented with effective prevention measures. It is also possible to save lives by detecting cancer earlier through screening when treatment is more likely to be successful. We believe that better methods to predict cancer can improve both cancer prevention and screening.
Aims:
We aim to develop a method that predicts cancer of any type.
Public health impact:
This project will provide a risk prediction model that can inform individuals their risk of developing any common cancer. It will allow individuals to understand the most effective ways they can reduce their risk, and also highlight if they may benefit from screening.
Expected Project duration:
3 years


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comprehensive-risk-factor-screening-and-genetic-susceptibility-loci-for-non-communicable-diseases

Comprehensive risk factor screening and genetic susceptibility loci for non-communicable diseases

Last updated:
ID:
99157
Start date:
15 May 2023
Project status:
Current
Principal investigator:
Mr Yi Feng
Lead institution:
Guangzhou Medical University, China

Aims!
Our goals are as follows: 1) To analyze the risk factors of non-communicable diseases (cancer, cardiovascular and cerebrovascular diseases, diabetes and other system-related diseases) using the large prospective database of UK Biobank, so as to determine the risk factors and facilitate the prevention of the corresponding exposure factors. 2) Determine genetic susceptibility locus according to the corresponding risk factors, and accurately guide individual prevention and clinical application through comprehensive application.

Scientific Rationale!
With the growth of life expectancy, the trend of population aging is becoming more and more obvious. Meanwhile, the incidence of related non-communicable diseases (cancer, cardiovascular and cerebrovascular diseases, diabetes and other system-related diseases) is on the rise. In this context, it is particularly valuable to carry out research on non-communicable diseases. It is now known that complex diseases such as non-communicable diseases are influenced by genes and environment. For example, not everyone who smokes are susceptible to lung cancer. One possible explanation is that some people may be born with genetic variants that makes them more susceptible to lung cancer if they smoke. The influence of the environment, such as air pollution and occupational exposure, has been identified as the risk factors of diseases. In addition, many lifestyles, drug history, and previous medical history may also have disease risks, so it is necessary to comprehensively analyze the influence of exposure factors on non-communicable diseases. We want to use the large prospective study of UK Biobank to conduct comprehensive risk factor screening and genetic susceptibility locus identification for non-communicable diseases.

Project duration: 36 months

Public health impact:
The identified risk factors can provide preventive advice to the population and reduce the occurrence of diseases, thus reducing the huge medical economic burden caused by non-communicable diseases. The identification of genetic susceptibility locus and polygenic risk scores can indicate the individual’s risk of developing corresponding non-communicable diseases and exert clinical application.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comprehensive-risk-factor-screening-and-genetic-susceptibility-loci-for-respiratory-system-diseases

Comprehensive risk factor screening and genetic susceptibility loci for respiratory system diseases

Last updated:
ID:
749045
Start date:
23 July 2025
Project status:
Current
Principal investigator:
Dr Qixia Wang
Lead institution:
Guangzhou Medical University, China

Research Questions:
1. What are the key demographic, lifestyle, environmental, and metabolic risk factors associated with respiratory diseases?
2. Which genetic loci are linked to respiratory disease susceptibility?
3. How do genetic and environmental factors interact to influence disease risk?
Objectives:
1. Utilize the UK Biobank dataset to systematically analyze risk factors for respiratory diseases, including COPD, asthma, bronchiectasis, interstitial lung disease, pneumonia, and lung cancer.
2. Identify genetic susceptibility loci through genome-wide association studies (GWAS) and assess cumulative genetic risk using polygenic risk scores (PRS).
3. Investigate gene-environment interactions to understand how genetic predisposition modifies the effect of external risk factors.
Scientific Rationale:
Respiratory diseases pose a growing burden on healthcare systems, yet their risk factors and genetic underpinnings remain incompletely understood. While environmental exposures such as smoking and air pollution are well-established contributors, genetic predisposition plays a crucial role in disease susceptibility. However, the complex interplay between genetic and environmental factors is not fully elucidated. By integrating comprehensive epidemiological and genomic data from the UK Biobank, this study aims to improve disease prediction, enable targeted prevention strategies, and advance precision medicine approaches. Findings will support public health interventions and inform personalized risk assessment for respiratory diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comprehensive-study-of-breast-cancer

Comprehensive Study of Breast Cancer

Last updated:
ID:
139541
Start date:
5 November 2024
Project status:
Current
Principal investigator:
Dr Zhiyang Li
Lead institution:
Shantou University, China

Breast cancer is the most common malignant tumor in the world, and it is also the leading cause of cancer death in women worldwide, and its mortality rate ranks fifth. Although breast cancer has made significant progress in risk factors, early diagnosis and treatment strategies, especially improvements in chemotherapy, endocrine therapy, targeted therapy and immunotherapy, its prognosis is still not ideal. Therefore, as a relatively comprehensive study, our project will last for a long time, at least 3 years. We hope to use database-related breast cancer data sets and use some statistical methods, R language, machine learning and other means to explore more Regarding the pathogenesis, potential targets and related prognostic factors of breast cancer, it will provide more reference for further guiding reasonable and standardized individualized treatment, and make a great contribution to improving the prognosis and quality of life of breast cancer patients.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comprehensive-study-of-genome-phenome-associations-in-complex-diseases-and-comorbidities-in-multi-ethnic-populations

Comprehensive study of genome-phenome associations in complex diseases and comorbidities in multi-ethnic populations

Last updated:
ID:
46341
Start date:
2 October 2019
Project status:
Current
Principal investigator:
Professor Bonnie Berger
Lead institution:
Broad Institute, United States of America

Clinical medicine has long recognized that certain diseases often occur together. For example, many individuals with autism spectrum disorder also have inflammatory bowel disease. Unfortunately, the shared genetic and molecular mechanisms that underlie these connections are poorly understood. Clinicians are therefore forced to treat each symptom individually instead of treating the shared pathology. The search for treatments can thus be time-consuming and frustrating for both patients and medical providers as relief from symptoms may be elusive. This is further complicated by the fact that a person’s individual response to treatment depends on their unique set of genetic and environmental risk factors. Yet, how diverse ethnic backgrounds and environmental exposures affect diseases is also poorly understood.

The aim of this study is to help fill both of these knowledge gaps. Our goals are 1) to uncover the genetic and molecular commonalities which cause certain diseases to occur together and 2) to understand how these risk factors are influenced by ethnicity and environment. To do so, we will apply the latest in machine learning and artificial intelligence to understand these patterns using the diverse data available through the UK Biobank. We will report our findings in a way that visually summarizes the connections between diseases and their genetic and molecular mechanisms, connections between patients’ symptoms and genetic makeup, and connections between genetic makeup and response to treatments. We believe our findings will help streamline the healthcare process, moving clinical practice towards medicine personalized to each person.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/comprehensive-study-of-mosaic-loss-of-chromosome-y-in-aging-males-and-predisposition-to-cancer-as-well-as-alzheimers-disease-using-the-uk-biobank-cohort

Comprehensive study of mosaic loss of chromosome Y in aging males and predisposition to cancer as well as Alzheimer’s disease using the UK Biobank cohort.

Last updated:
ID:
13135
Start date:
1 October 2015
Project status:
Current
Principal investigator:
Professor Jan Dumanski
Lead institution:
Uppsala University, Sweden

We have recently made three discoveries related to acquired Loss Of chromosome Y (LOY) in blood cells of aging men. LOY is associated with: a) shorter survival and increased mortality from cancers; b) Alzheimer?s disease (AD); and c) smoking of tobacco, suggesting that smoking induces LOY. These findings are a basis for research towards understanding why LOY causes cancer and AD in males.

Our aim is to further explore LOY as a disease biomarker in ~250,000 males of UK Biobank and (using the female cohort) investigate chromosome X to search for corresponding mechanisms that operate in women. The questions, which are the focus of our application, are clearly of public health importance. The results may significantly improve our understanding of the risks for cancer, Alzheimer?s disease and perhaps other morbidities in men, and possibly also in women. Cancer and Alzheimer?s disease diagnoses are relevant for >50% of morbidity and/or mortality in aging human population. Our project could also be critical in identifying an extreme at-risk subgroup for beneficial health interventions. We will use Axiom data to measure LOY in males. This information is not available from standard output and requires intensity data from the Axiom genotyping.
We will also correlate LOY with smoking, mortality, cancer, Alzheimer?s disease (AD) and other outcomes. In parallel, we will investigate whether chromosome aberrations occurs in the blood of women, by looking for loss (full or partial) of the X chromosome in Axiom data. Full cohort for Axiom data and health information will be required.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/compressed-sensing-and-high-dimensional-statistical-methods-in-complex-trait-genomics

Compressed Sensing and high-dimensional statistical methods in complex trait genomics

Last updated:
ID:
15326
Start date:
3 October 2015
Project status:
Current
Principal investigator:
Professor Stephen Hsu
Lead institution:
Michigan State University, United States of America

Our goal is to test new computational methods for determining the genetic architecture of complex traits, including highly heritable conditions such as Type 1 Diabetes, Alzheimer’s, and others. The techniques we plan to use have been the subject of intense recent activity in fields such as optimization, signal processing and machine learning, but so far have just begun to be applied in genomics. The research will produce improved predictive models which, based on individual genomics, identify individuals at high risk for certain diseases. It will also identify the many alleles associated with this risk. Early intervention with high risk individuals may decrease rates of incidence and reduce health care costs. Elaboration of underlying genetic architecture is important basic science and may lead to improved treatments (e.g., drug development). We wish to obtain access to genomic data and phenotype data relevant to highly heritable disease conditions (e.g., Type 1 Diabetes) as well as complex traits such as height, BMI, cognitive ability. Advanced computational algorithms will be used to study the genetic architecture of these traits. The techniques we plan to use have been the subject of intense recent activity in fields such as optimization, signal processing and machine learning, but so far have just begun to be applied in genomics. Analysis will be performed on high-performance computing clusters. We would like access to the full cohort (SNP genotypes), and several relevant phenotypes.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/computational-analyses-of-genotypic-and-phenotypic-data-with-treatment-response-from-patients-with-multimorbidity-and-the-role-of-inflammation-as-a-driver-of-multimorbidity

Computational analyses of genotypic and phenotypic data with treatment response from patients with multimorbidity and the role of inflammation as a driver of multimorbidity.

Last updated:
ID:
48433
Start date:
18 June 2019
Project status:
Current
Principal investigator:
Dr Priyank Shukla
Lead institution:
Ulster University, Great Britain

Hospitals in UK see 40-50% older patient, who are experiencing shorter life expectancy and increasing number of severities, which starts to affect their function and quality of healthy life years. Such patients suffer from multiple chronic conditions (multimorbidity). Current clinical care of multi-morbidities is based largely on the guidelines for treating the single diseases separately, and patients with multimorbidity are frequently excluded from clinical trials. We hypothesize that stratification can aid disease management and significantly improve the present poorly served clinical practices for multimorbid patients. Also, most of the current therapies don’t address the nexus between inflammation and multiple chronic conditions in a patient. To tailor the best therapy, a trade-off between mortality risk and disability risk must be mutually agreed between patient and therapist. As a result, we would like to come up with risk score-based strategies to achieve the same.

Machine Learning (ML) based algorithms have been widely used for prediction/classification problems in bioinformatics. With the continued deployment of advanced high-throughput omics technologies (specially NGS) in clinical practice, AI offers significant opportunities to assist with the analysis of the terabytes of clinical and omics data being generated from patients.

The project will require 6 months of data pre-processing, followed by 2 years of rigorous application of AI/ML computational techniques and finally 6 months of collation of results.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/computational-and-machine-learning-tools-for-the-analysis-of-genetic-and-medical-imaging-data-to-study-human-health-and-disease

Computational and machine learning tools for the analysis of genetic and medical imaging data to study human health and disease

Last updated:
ID:
87065
Start date:
13 June 2022
Project status:
Current
Principal investigator:
Dr Francesco Paolo Casale
Lead institution:
Helmholtz Zentrum Munchen, Germany

The availability of medical imaging data in large genetic cohorts enables the study of human health and disease at an unprecedented scale and resolution. For example, these datasets can help identify new quantitative imaging biomarkers of disease, characterise the associated pathological processes and identify their driving genes and pathways through genetic analyses. Despite their great promises, joint analyses of genetic and medical imaging data still present several computational and interpretation challenges and general frameworks for these analyses are not fully established.

In this project, we aim to develop computational tools to extract complex phenotypic patterns from medical imaging data, identify their genetic and environmental drivers, and establish their link with human health and disease. To do so, we will extend and combine tools from the fields of deep learning, statistical genetics and causal inference. We propose to use the UK Biobank resource to benchmark these tools across different imaging modalities and indications, including but not limited to brain MRIs for degenerative and neurological disorders, abdominal MRI for metabolic disorders, cardiac MRIs for different cardiovascular diseases and conditions, and DXAs for osteoarthritis. Considering that our project involves the analysis of multiple imaging modalities and the development of general computational tools, we expect it to take several years. Nevertheless, a successful outcome of this project can lead to new disease biomarkers usable in the clinic and insights that could lead to the development of new therapeutics.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/computational-approaches-to-understanding-the-full-spectrum-of-variant-effects-in-rare-disease-and-common-neurological-traits

Computational approaches to understanding the full spectrum of variant effects in rare disease and common neurological traits.

Last updated:
ID:
81050
Start date:
14 September 2022
Project status:
Current
Principal investigator:
Dr Nicola Whiffin
Lead institution:
University of Oxford, Great Britain

Knowing the precise genetic change that causes disease in an individual is of huge importance to both a patient and their family. This enables screening of other family members to identify those also at risk, prenatal screening, and can lead to personalised treatments. Current approaches to search for these causative genetic variants (or ‘genetic diagnoses’) are only successful in around half of all individuals with a rare disease. These approaches focus only on regions of DNA that directly encode proteins, however, these regions only comprise ~1.5% of the total genomic DNA. Our research aims to increase our understanding of variants in other regions of the genome, so called ‘regulatory regions’ that modify the amount of proteins that are produced, when they are produced, and where they are located in the cell. Through this work we aim to increase the ‘search space’ for genetic diagnoses and hence enable a causative variant to be identified in more individuals with rare disease.

In another branch of research we are interested in two common neurological traits that affect social perception and memory. The first of these is prosopagnosia, or face-blindness, which is the inability to recognise faces. The other is aphantasia, which is a complete absence of mental imagery (i.e. being unable to either form or recall images in the mind). Both of these traits are common, occurring in ~2-5% of the population, however, we know very little about the genetic variants that influence these traits. Our research will uncover genetic variants that are involved in these traits, enabling us to discover more about the underlying biology and connections to other neurological conditions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/computational-modelling-of-human-heart-using-mr-images

Computational Modelling of Human Heart using MR Images

Last updated:
ID:
32263
Start date:
1 September 2017
Project status:
Closed
Principal investigator:
Jayendra Bhalodiya
Lead institution:
University of Warwick, Great Britain

Human heart MR Image analysis to develop an automatic procedure for cardiovascular diseases diagnosis and prognosis. The patients suffering from hypertrophic and dilated cardiomyopathy, arrhythmia, ischemia can be benefitted with improved desease identification and futture state estimation. The research question targetted is to develop algorithms for cardiac motion tracking and image registration to estimate muscles deformation and strain values in healthy as well as diseased heart patients. Heart muscles related diseases have been addressed to develop fast and accurate diagnosis. The calculated results will be compared with different clinical measures addressing the research question of accuracy of various clinical measures. The human heart muscles myocardial properties have been addressed in this research to improve diagnosis and surgical plan. The outcome will help patients of cardiovascular disease like arrhythmia, ischemia, dilated and hypertrophic cardiomyopathy that is a leading cause of death globally. Therefore the purpose of UK Biobank to improve the health of humans by developing an advanced technological tool to accurately analyse and treat diseases will be achieved. The research will be simulation and algorithm based using anonymised MR Images at University of Warwick. There will no be any involvement of patients. The research will be to develop algorithms to analyse medical images using mathematical models. Computer algorithms with the mathematical framework will be used to analyse data cohort restricted to heart imaging. There will not be any participants involved. The anonymised MR Image data that is available in UK Biobank will be used for research. We require a cohort restricted to those with heart imaging data.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/computational-phenotyping-to-understand-disease-progression-across-the-human-lifespan

Computational phenotyping to understand disease progression across the human lifespan

Last updated:
ID:
82779
Start date:
24 May 2022
Project status:
Current
Principal investigator:
Dr Nicola Pirastu
Lead institution:
Fondazione Human Technopole, Italy

In the last century improvement in general societal conditions and technological advance in medical care has increased life expectancy dramatically. This has resulted in longer lives but also lengthening the proportion of life which is characterised by chronic health conditions. It is thus a fundamental goal for health research today to not only further increase life expectancy, but to improve quality of health and thus life quality in the last part of our lives. This goal needs much better prevention interventions and medications to be achieved, which require a better understanding of both the biological and non-biological factors which lead to a decreased health status before the onset of disease.
UK Biobank is unique in the sense that is had deeply phenotyped individuals (thousands of recorded measurements, diagnoses, medications, etc) and large-scale multi-modal imaging (MRIs, retinal fundus scans, Optical coherence tomography, etc). Imaging data is a very complicated and rich source of information which hasn’t been fully utilised to date. We will develop machine learning algorithms to automatically extract phenotypes from these images, that can help us characterise the chronic conditions of ageing.
Our project has the bold aim of combining statistical and bioinformatics approaches to dissect the complex relationships which lead to a reduced quality of life, understanding which factors are driving complex disease burden and how. Furthermore, a primary focus will be to examine the role that inflammation plays in the healthy aging process and how this relates to the measures derived from the imaging data.

Initially, we set the duration of the project at 36 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/computational-strategies-for-translational-pan-genomics

Computational strategies for translational pan-genomics

Last updated:
ID:
304181
Start date:
28 May 2025
Project status:
Current
Principal investigator:
Professor Alexander Schönhuth
Lead institution:
Bielefeld University, Germany

As of today, the volume of available genome data has reached a
critical mass, enabling its exploitation through advanced artificial
intelligence (AI) approaches. Through corresponding analyses, crucial
individual genetic variations can be pinpointed, supporting clinical
diagnoses and facilitating the determination of appropriate treatment
protocols with an unprecedented level of accuracy in the history of
medicine.

However, effectively harnessing these data masses necessitates
advanced techniques for their analysis. Here, we propose organizing
the data using methods from “computational pan-genomics,” an area of
research focused on the efficient and compact arrangement of genomes
from entire populations. This involves arranging genomes in a manner
that is both efficient and compact. Additionally, we aim to exploit
the (evolutionarily/genetically coherent) organized data within
advanced AI frameworks.

In pursuit of these objectives, we encounter two specific challenges.

Firstly, we aim to leverage the fundamental knowledge gleaned from
large, disease-unspecific masses of genomes for the targeted analysis
of rarer diseases. This strategy involves two steps. Initially, one
learns everything possible about (evolutionarily related) genomes in
general, commonly referred to as “pre-training.” Subsequently, the
focus shifts to the specific rare disease of interest, commonly known
as “fine-tuning.” Despite the scarcity of data for rarer diseases,
various recent protocols demonstrate the success of this step-wise
strategy. Pursuing such strategies will help mitigate biases that
hinder the study of rare diseases, as more frequent diseases often
receive disproportionate attention, potentially overshadowing research
on rarer conditions.

Our second objective is to devise knowledge exploitation strategies
that safeguard the privacy of individuals contributing their genome
records for general usage. Publishing successful privacy-preserving
strategies will encourage individuals to make their genome data
accessible to researchers.

Both goals rely on the latest advances in computational pan-genomics
and advanced machine learning. The integration of these two domains is
commonly referred to as “translational pan-genomics.” We are confident
that the synergy between massive data, advanced computational
pan-genomics data organization techniques, and advanced AI frameworks,
such as large language models, holds breakthrough potential in the
near to mid-term future.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/computer-assisted-study-of-regulatory-gene-variants-expression-implicated-in-type-2-diabetes-and-various-comorbid-diseases-in-the-european-populations-of-russia

Computer-assisted study of regulatory gene variants expression implicated in type 2 diabetes and various comorbid diseases in the European populations of Russia.

Last updated:
ID:
59397
Start date:
27 July 2020
Project status:
Closed
Principal investigator:
Mr Vasiliy Reshetnikov
Lead institution:
The Institute of Cytology and Genetics SBRAS, Russian Federation

The International Diabetes Federation estimated that 415 million adults had diabetes mellitus in 2015. Major risk factors in the development of this disease and its complications are obesity, lifestyle factors, genetic predispositions, epigenetics and early developmental factors. Notwithstanding huge amount of papers demonstrating the influence of many genetic variants on type 2 diabetes development, there are numerous conflicting results that cast doubt on the molecular-genetic basis determining the development of the disease.
The aim of the proposed study is to elucidate the genetic and epigenetic causes of type 2 diabetes associated with regulatory gene variants, that resides in enchancers and promoters, worldwide and in particular, in Russia. In order to do this, we should answer the numerous questions. The main one is: What is the molecular basis of phenotypical penetration and expression of heterozygous regulatory and coding gene variants implicated in type 2 diabetes and comorbid diseases?
On the one hand, Onuchic et al. (doi: 10.1126/science.aar3146) revealed sequence-dependent CpG methylation imbalances at thousands of heterozygous regulatory loci in human cells that are enriched for random transitions between fully methylated and unmethylated states of DNA, which in turn could be the main disease-associated factors. On the other hand, a recent breakthrough in identification biomarkers of aging based on DNA methylation data (doi: 10.1038/s41576-018-0004-3; doi: 10.1093/nar/gkx1139) links organism development and cell identity maintenance to biological aging and various age-dependent diseases. Hence, it is tempting to analyze the bulk of data of this CpG methylation in-depth in order to calculate scores (such as polygenic risk scores) that help physicians to give an early diagnosis and treat diseases based on in-depth knowledge on epigenetic dynamics in heterozogous loci.
In our proposed work for analyzing the phenotypical expression of heterozygous regulatory gene variants implicated in type 2 diabetes we will construct various genetic context-driven, phenotypic-awared and cohort-informed prioritized or weighted datasets. After this, we will analyze and annotate these datasets by various available statistical procedures and with all known databases depositing genome-wide data to uncover heterozygous regions that characterizes by high cumulative association between the ranks of risk (and/or negative prognosis) of type 2 diabetes (and/or comorbid diseases) and special signatures in genetic context.
We plan to complete the project within one and half year. After this time period, we will construct screening panels composed of loci guiding physicians to reach an early diagnosis and treat type 2 diabetes and comorbid diseases in a more effective way.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/concurrent-challenges-prognosis-of-pulmonary-hypertension-in-lung-cancer-patients

Concurrent Challenges: Prognosis of Pulmonary Hypertension in Lung Cancer Patients

Last updated:
ID:
193312
Start date:
26 March 2024
Project status:
Current
Principal investigator:
Dr Fang Zhu
Lead institution:
Shanghai Jiao Tong University School of Medicine., China

Title: Concurrent Challenges: Prognosis of Pulmonary Hypertension in Lung Cancer Patients

Aims and Scientific Rationale:
Our research aims to explore a crucial health issue: the impact of pulmonary hypertension (PH) on patients with lung cancer. Lung cancer is a leading cause of cancer death worldwide, and its prognosis is often worsened when combined with PH, a condition characterized by high blood pressure in the lungs. This study seeks to understand how PH affects lung cancer outcomes, the role of PH severity in lung cancer treatment, and to identify any common characteristics and genetic markers in lung cancer patients who develop PH. Our goal is to improve treatment and management strategies for these patients.

Project Duration:
From Jan 2024 to Dec 2029

Public Health Impact:
This research holds significant public health importance. By understanding the interplay between lung cancer and PH, we can develop better screening methods, leading to earlier diagnosis and potentially more effective treatment options. Additionally, by identifying genetic markers associated with worse outcomes in lung cancer patients with PH, we can move towards more personalized medicine. This means treatments can be tailored to individual patients based on their genetic makeup, improving the effectiveness of the treatment and potentially reducing side effects.

Our findings could also guide healthcare providers in making informed decisions about managing lung cancer patients who also suffer from PH. This is crucial, as managing one condition without considering the other could lead to less effective treatment and poorer patient outcomes. Furthermore, our research might influence health policy and education, ensuring resources are allocated efficiently and healthcare professionals are well-informed about this complex medical issue.

In summary, this study not only aims to fill a gap in current medical research but also has the potential to significantly improve the lives of lung cancer patients with PH. It could lead to advancements in treatment, better patient care, and ultimately, contribute to reducing the burden of lung cancer and PH on patients and healthcare systems worldwide.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/confirmation-and-expansion-of-nih-intramural-results-related-to-brain-imaging-gene-dose-effects-and-genetic-scores

Confirmation and expansion of NIH intramural results related to brain imaging, gene-dose effects and genetic scores

Last updated:
ID:
22875
Start date:
22 February 2018
Project status:
Current
Principal investigator:
Dr Adam Thomas
Lead institution:
National Institute of Mental Health, United States of America

Findings we would like to confirm include the association between gyrification and general cognitive ability (Gregory et al., 2016), allometric analysis of the relationship between sex chromosomes and cerebellar organization and subcortical anatomy (Reardon et al., 2016; Mankiw et al. 2017), and association between genome-wide copy number variations and brain morphometry and connectivity (Elia et al., 2011).

The research will aid in establishing relationship between brain imaging and phenotypic data. We will apply existing image processing software, including the AFNI software package developed at the NIH, for analysing the imaging data being acquired by Biobank. AFNI provides tools to perform image alignment, skull stripping, and statistical processing that is similar to those previously used on the Biobank UK imaging data. This independent processing stream will provide a valuable opportunity to compare and validate the different processing tools against one another. We will repeat genetic analyses conducted in our existing publications to confirm previous findings and look for novel findings with the assistance of the larger datasets.
We will use all of the available participants with brain imaging data which will build to 100,000 participants.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/connecting-environment-to-health-through-dna-methylation

Connecting Environment to Health through DNA methylation

Last updated:
ID:
966158
Start date:
28 September 2025
Project status:
Current
Principal investigator:
Professor Jason Buenrostro
Lead institution:
Broad Institute, United States of America

The Epigenomics Program and the Biology of Adversity Project at the Broad Institute are looking to discover biomarkers of exposure (e.g. stress, lifestyle, environmental exposure, healthy history etc) that predispose individuals to disease and reduced health span. The biological pathways linking long-term stress exposure to disease onset remain poorly defined. We aim to leverage UK Biobank data to identify robust, biologically meaningful biomarkers of exposure that can elucidate mechanisms of disease vulnerability. This research will integrate diverse data modalities, with three areas of focus. First, we will harness the UK Biobank’s extensive and diverse participant metadata to examine how variables such as socioeconomic status, early-life adversity, lifestyle factors, and ethnicity intersect with exposure and disease trajectories. This approach ensures inclusivity and improves the generalizability of biomarker discovery across populations. Second, we will utilize high-resolution proteomic data (including Olink and plasma proteomics) to investigate circulating peptide hormones involved in the hypothalamic-pituitary-adrenal (HPA) axis. Particular attention will be given to hormones such as cortisol, ACTH, and CRH, which are central to the stress response. By correlating hormone profiles with stress exposure histories, we aim to identify proteomic signatures reflective of chronic HPA axis activation. Third, we will analyze genome-wide DNA methylation patterns to detect gene-specific epigenetic modifications associated with exposure. Methylation signatures will be mapped to genes involved in stress signaling, immune response, and metabolic regulation, allowing us to explore how environmental stressors become biologically embedded and influence gene expression. These efforts will reveal biomarkers spanning social, proteomic, and epigenetic domains that mediate the effects of chronic stress on health, paving the way for early detection tools and preventive strategies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/connecting-properties-of-the-micro-and-macro-vasculature-from-multimodal-imaging-through-genetics-and-deep-learning-to-better-understand-vascular-pathomechanisms-and-predict-disease-risk

Connecting properties of the micro- and macro-vasculature from multimodal imaging through genetics and deep learning to better understand vascular pathomechanisms and predict disease risk

Last updated:
ID:
90947
Start date:
27 October 2022
Project status:
Current
Principal investigator:
Professor Sven Bergmann
Lead institution:
University of Lausanne, Switzerland

Each year cardiovascular diseases (CVD) cause 3.9 million deaths in Europe, amounting to 45% of all deaths. While certain risk factors like age, smoking and hypertension have been well documented, the impact of blood vessel characteristics is poorly understood. Vascular properties – such as shape and size of the blood vessels – can be investigated with non-invasive and inexpensive imaging methods in some organs, such as the retina. For example, the bendiness (also known as “tortuosity”) of retinal vessels has been shown to be associated with increased risk of CVD. However, the extent to which vascular properties extracted from retinal images reflect those of other body parts has not been studied systematically.
Our project aims at providing an extensive analysis of vascular properties of the retina and link them to those from other organs, such as the brain or the heart. We will use genetic information about such properties to investigate the biological mechanisms that link vascular properties to CVD. Using machine learning, we will test whether we can predict CVD risk in the general population, based on vascular properties derived from multi-organ imaging and genetic information.
Our findings have the potential to increase our understanding of the pathological mechanisms leading to CVD and provide tools for the detection of presymptomatic vascular modifications that can support early diagnosis of vascular diseases. We propose to complete this project in 36 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/connecting-rejuvenation-therapies-to-age-associated-human-disease-indications

Connecting rejuvenation therapies to age-associated human disease indications

Last updated:
ID:
101888
Start date:
26 May 2023
Project status:
Current
Principal investigator:
Dr Christian Hammer
Lead institution:
Altos Labs, Inc., United States of America

Diseases of ageing present a large unmet clinical need across the world. We will use data from the UK Biobank to study how the rate of ageing differs between individuals, identify how the DNA variants that drive these differences act to change the molecular features of our cells (such as differences in the genes that are expressed), and how this impacts on human phenotypes, such as disease. This may help us to develop therapeutics that are targeted to the diseases where they are most likely to be successful.

We will also use the UK Biobank data to identify individuals (whether by genetic background, biomarker levels, MRI scans, or medical record data) that are more likely to respond to a particular therapeutic approach and/or that are at greater risk of a particular disease or other phenotype. We will use the entire cohort of the UK Biobank in order to maximise our ability to detect these effects.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/consequences-of-low-cholesterol-syndromes-due-to-truncated-apob-or-mtp-and-angptl3-inactivation-on-chronic-liver-and-cardiovascular-diseases-a-mendelian-randomization-study

Consequences of low cholesterol syndromes due to truncated ApoB or MTP and ANGPTL3 inactivation on chronic liver and cardiovascular diseases: a Mendelian randomization study.

Last updated:
ID:
70790
Start date:
28 January 2022
Project status:
Current
Principal investigator:
Professor Marcello Arca
Lead institution:
Sapienza Universita di Roma, Italy

New medications to lower cholesterol in the blood have been developed. These new therapies are able to inhibit the production of a lipoprotein called very low-density lipoprotein (VLDL), which is made by the liver and carry triglycerides (TG). This effect is obtained by blocking three proteins called microsomal transfer protein (MTP), apolipoprotein B (ApoB) and angiopoietin like 3 protein (ANGPTL3). The MTP protein is responsible for putting together TG and ApoB to form the VLDL. The ANGPTL3 protein blocks the degradation of VLDL so that getting it out of the way we may expect to accelerate the elimination of VLDL.
Several studies in humans have demonstrated that these new drugs can bring plasma cholesterol and triglycerides at very low levels, which is a good thing for the people who must be cured because they have high lipids in the blood. However, one potential problem we may encounter if we block these proteins, is that people might accumulate TG in the liver. This condition is called hepatic steatosis and can predispose to more severe diseases, such as liver cirrhosis or liver cancer.
Three rare genetic disorders due to mutations in genes that reduce the production of MTP, ApoB and ANGPTL3 proteins can be taken as examples of what can happen to the liver if we block these proteins. Indeed, individuals with mutations in MTTP and APOB genes are prone to develop steatosis, while those with mutations in ANGPTL3 did not show any fat in the liver. In any case, patients affected by these rare diseases appear to be naturally protected against cardiovascular disease (CVD), probably due to the strong reduction of blood cholesterol.
Then, in order to elucidate the possible harmful effects on the liver of pharmacological blockage of MTP, ApoB, and ANGPTL3 proteins, we have planned to employ the experimental procedure called Mendelian Randomization. Following this procedure, we aim to compare the health status of the liver in individuals carrying mutations in APOB, MTTP, and ANGPTL3 genes vs. those not carrying any mutations. As a secondary objective, we will evaluate whether these mutations may determine a reduced risk of CVD. To achieve this aim, we will need 3-years. Our results may be useful for guiding the safety monitoring of the new upcoming cholesterol-lowering drugs.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/construct-a-composite-wellbeing-measurement-and-resilience-scale-in-uk-biobank-for-use-in-gwas-on-wellbeing-and-resilience

Construct a composite wellbeing measurement and resilience scale in UK-biobank, for use in GWAS on wellbeing and resilience

Last updated:
ID:
58534
Start date:
18 May 2020
Project status:
Closed
Principal investigator:
Mr Javad Jamshidi
Lead institution:
University of New South Wales, Australia

This research has three main aims:
1. To construct a quantitative measurement, using multiple mental health questions, to capture a broad concept of wellbeing and perform a GWAS using the wellbeing score.
2. To define a quantitative score for resilience for people exposed to trauma, based on the composite wellbeing score, and not only the absence of mental illness such as PTSD. Then the conduct of a GWAS based on this quantitative measure of resilience.
3. To investigate the possible correlation between the grey matter volume of different brain structures and the wellbeing and resilience score.
Although wellbeing seems to be a simple concept, it has different facets and there are various instruments to measure different aspects of it. Wellbeing is a spectrum, and all its components correlate with each other. Therefore, it is better to consider it as a whole, and include all its component aspects, when trying to measure it. In this project, we are trying to make a composite instrument using the questions available in the UK Biobank in order to capture a broader definition of wellbeing, which includes all facets of wellbeing, then perform a GWAS to reveal the genetic variations that influence it. This approach will help us to have a better perspective on wellbeing and identify the factors that have the most influence on our mental health and wellbeing. Defining resilience using wellbeing itself (as a spectrum) rather than just presence or absence of mental illness, will be instrumental in understanding factors which make us resilient or vulnerable to trauma. It will help us to understand the factors which make us more susceptible to mental illness, while we are still on the so-called normal spectrum of wellbeing or mental health.
This project will take ~16 months to complete after the data is provided. Six months for construction of the wellbeing instrument and related analysis, about 6 months for doing the genetic analysis and 4 months for the imaging study.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/constructing-a-diagnostic-prediction-model-for-attention-deficit-hyperactivity-disorder

Constructing a diagnostic-prediction model for Attention-Deficit Hyperactivity Disorder

Last updated:
ID:
85636
Start date:
22 August 2022
Project status:
Current
Principal investigator:
Professor Takashi Makino
Lead institution:
Tohoku University, Japan

Attention-Deficit/Hyperactivity Disorder (ADHD) is a neurodevelopmental disorder with a prevalence of about 2.5% in adults and 5% in school-aged children. Twin studies have shown that the disorder is inherited with a probability of about 70%. The current diagnostic criteria for ADHD are mostly subjective, and the diagnosis is made without the use of genetic biomarkers, even though previous studies have strongly suggested that the disorder is hereditary. The results of the genome-wide association study did not identify any genes with large effect sizes that are closely related to the expression mechanism of ADHD, as reported in previous studies. This suggests that many genetic factors with small effects are involved in ADHD, and that the phenotype of ADHD is not manifested only by specific genes. Therefore, we are conducting research activities to construct a diagnostic prediction model of ADHD based on genome-wide genetic information by utilizing machine learning and other methods with many candidate genetic causes reported so far to be associated with ADHD. The purpose of this study is to overcome the problem that ADHD has ambiguous criteria for diagnosis, and to construct a predictive model that assists the diagnosis of ADHD with objective indicators. The project period is 24 months.
As described in A6, it is expected that the diagnosis of psychiatric disorders, including ADHD, can be made objectively using genetic and biological indicators.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/constructing-a-genetic-effect-network-to-predict-polygenic-disease-risk

Constructing a genetic effect network to predict polygenic disease risk

Last updated:
ID:
60468
Start date:
29 May 2020
Project status:
Closed
Principal investigator:
Mr Austin Conklin
Lead institution:
University of Arizona, United States of America

Coronary artery disease (the primary cause of heart attack) is heritable. Currently, geneticists are working on methods to predict whether or not someone will develop coronary artery disease based on that person’s genetic makeup. We aim to improve on those methods using data from a large study of human tissues. The study measures gene “expression” (how active a certain gene is) in a given tissue and determines which genetic variants cause changes in expression. An import finding from this study (and from many other such studies) is that a genetic variant changes gene expression differently depending on which tissue you measure its effect in. We aim to take this tissue-specific manifestation data and use it to improve prediction scores. The rationale for this is that some genetic variants have a big effect on some tissues but not others; current risk predictors in the field are agnostic to this fact. We believe that combining tissue-specific genetic effects and risk prediction will allow us to predict a person’s coronary artery disease risk more accurately. That way, people at high risk can be prescribed drugs to mitigate the development of heart disease. We plan to work on this project for the next two years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/constructing-an-aging-clock-with-multi-omics-data-a-comprehensive-analysis

Constructing an Aging Clock with Multi-omics Data: A Comprehensive Analysis

Last updated:
ID:
584010
Start date:
5 April 2025
Project status:
Current
Principal investigator:
Dr Zhaoxiang Wang
Lead institution:
Zhejiang Lab, China

Aging is an inevitable physiological process that shows signs of functional decline at the tissue and cellular levels over time. Chronic diseases such as neurodegenerative diseases, cardiovascular diseases, metabolic disorders, and immune system dysfunction have become the main threat to the decline of quality of life and the end of life of the elderly. Therefore, it is particularly urgent to seek strategies and biomarkers to delay aging, and to develop early personalized interventions. We will focus on the following aspects:
1. Construction of aging prediction model
Integrating the biological datasets-genomics, proteomics, metabolomics, imaging, and behavioral data-to build a comprehensive aging prediction model. The model is instrumental in forecasting individual biological age and pinpointing biomarkers with the potential to retard the aging process.
2. Study of the causal relationship between lifestyle and aging, and to propose personalized regulation strategies
To harness genomic data to perform a genome-wide association study (GWAS) coupled with Mendelian randomization analysis, thereby dissecting the intricate causal interplay between lifestyle, like physical activity, dietary habits, sleep patterns, and psychological well-being, with the biological age. The objective is to uncover the factors that may expedite the aging process, offering insights for crafting personalized, multifaceted intervention strategies, such as augmenting physical exercise, refining dietary intake, and fostering psychological resilience.
3. Mediation analysis of behaviour – biological age – chronic diseases
Using mediation analysis, where behavioral and psychological attributes are treated as independent variables, biological age as the mediator, and chronic diseases as the dependent variable, to elucidate the mechanisms through which biological age influences the onset and progression of chronic conditions linked to unhealthy lifestyle.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/constructing-genotype-and-phenotype-network-helps-reveal-phenome-wide-association-studies-in-biobanks

Constructing genotype and phenotype network helps reveal phenome-wide association studies in biobanks

Last updated:
ID:
102999
Start date:
18 April 2023
Project status:
Current
Principal investigator:
Miss Xuewei Cao
Lead institution:
Michigan Technological University, United States of America

n this project, we will develop new statistical methods to find genes responsible for complex human diseases and apply these methods to Biobank data sets. Over the last decade, genome-wide association studies (GWAS) have been widely performed to identify genetic associations for many complex diseases. A common limitation of GWAS is that they focus on only a single disease. Emerging evidence has shown that pleiotropy, the phenomenon of one genetic variant affects multiple diseases, is widespread in complex human diseases. Therefore, joint analysis of multiple diseases within a cohort presents an attractive alternative to single disease analyses and may provide new insights into the etiology of human diseases. Therefore, in this project, we plan to develop novel statistical methods to test the association between a genetic variant with a large number of diseases jointly. We plan to work on this project in the coming three years. We will publish the statistical methods, computational tools and biological findings from the data analysis and make the research results publicly available. Discovering novel genetic variants contributing to complex diseases may aid in improving diagnosis, treatment, and prevention, and have significant public health benefits.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/constructing-risk-prediction-models-of-non-communicable-diseases-based-on-multiple-risk-factors

Constructing risk prediction models of non-communicable diseases based on multiple risk factors

Last updated:
ID:
150543
Start date:
1 February 2024
Project status:
Current
Principal investigator:
Professor Cao Yanan
Lead institution:
Shanghai Jiao Tong University School of Medicine., China

Aims: The proposed project aims to clarify the key risk factors for non-communicable disease (NCD) and construct its risk prediction models based on genetic, behavioral and enviromental risk factors.

Scientific rationale: Over recent decades, rapid economic development and social advancements worldwide, as well as the lifestyle transition from nutrient deficiency to nutrient excess in developing regions have greatly increased the the morbidity and mortality of NCD, including diabetes, obesity, non-alcoholic fatty liver disease and cardiometabolic disease. To meet this challenge, the United Nations have set goals of reducing premature deaths due to NCD by one-third by 2030 to achieve a sustainable health development. In fact, the most effective and cost-saving strategy to successfully decrease the burden of NCD is to adopt population-wide preventive measures in the early stages of the disease. Therefore, clarify key risk factors and construct prediction models based on these contributors hold great promise to identify high risk populations, thus improve their overall healthy life expectancy and avoid unnecessary medical costs. Since NCD resulted from a variety of genetic, behavioral and environmental risk factors, the prediction model that integrates multiple risk factors (such as the genetic- environmental factors cooperative prediction model) has better predictive performance in theory. Therefore, this project plans to construct risk prediction models of NCD based on multiple risk factors through collecting multidimensional data of UK biobank.

Project duration: This project is expected to be completed in 36 months.

Public health impact: we believe the results of this project will effectively improve the risk stratification of NCD, optimize the effects of existing clinical care, and advance the development of translational medicine such as precise diagnosis and targeted therapy.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/constructing-risk-scores-of-longevity-dementia-and-related-disorders

Constructing risk scores of longevity, dementia, and related disorders

Last updated:
ID:
54520
Start date:
4 February 2020
Project status:
Closed
Principal investigator:
Dr Najaf Amin
Lead institution:
University of Oxford, Great Britain

In the context of global aging of the population, preventing non-communicable diseases – also referred to chronic diseases – is a major public health care challenge. Therefore, preventive medicine is gaining in interest. Over the last decade, enormous progress and advances have been achieved in the field of genetic research. These researches, coupled with other studies on non-genetic risk factors (i.e. lifestyle related factors), have led to the identification of high risk group of subjects. Thanks to targeted preventive campaigns and the modification of some risk factors, the incidence of most of chronic diseases has decreased in developed countries. However, major efforts still need to be made in order to better understand how the identified risk factors of one particular disease could influence the development of other diseases and/or affect longevity. For instance, patients with stroke and vascular disease show an increased cognitive decline and present an increased risk of dementia or Alzheimer’s disease. Here we aim to improve the risk scores by also including non-genetic risk factors including lifestyle related factors (e.g. education level, alcohol consumption, smoking history, history of hypertension, dyslipidemia, physical activity, body mass index (BMI)), or concomitant pathologies such as stroke, cardiovascular disease or chronic respiratory disease. Another important question to weigh in is the life expectancy of the individuals, both overall and in those at high risk of the disease under study.
The aim of this study is to construct lifetime risks for mortality and major late-onset disorders including neurodegenerative diseases, cardiometabolic and cerebrovascular diseases and respiratory diseases using genetic and non-genetic risk factors. We will study single disorders as well as comorbidity.
This project is expected to run over the course of 6 months.
In the context of global aging prevention of age related disorders such as dementia and cognitive impairment, depression has also been identified by the World Health Organization as a top priority. Insight into the lifetime risk of these disorders and the identification of high risk groups – genetic and non-genetic – is a of high urgency and will have a great impact on public health.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/construction-of-a-multi-omics-aging-assessment-model-based-on-biological-mechanisms-and-signal-pathway-interactions

Construction of a multi omics aging assessment model based on biological mechanisms and signal pathway interactions

Last updated:
ID:
778108
Start date:
29 August 2025
Project status:
Current
Principal investigator:
Dr Zhangjian Chen
Lead institution:
Peking University, China

Research Question: As organisms age, a series of changes occur in their epigenetics, proteins, and metabolites. How can we comprehensively evaluate human aging through the complex changes in epigenetics, proteins, and metabolites? What are the intrinsic connections between human chronological age and biological age, as reflected by the integrated indicators of epigenetics, proteins, and metabolites?

Research Objectives: (1) Utilizing multiple databases, we aim to conduct multi-omics joint analysis on healthy populations of different ages to screen for aging-related biomarkers and key signaling pathways. This will enable us to establish a signaling pathway-based model for assessing human aging. (2) We will calculate the general patterns and correlations between biological aging and chronological age at the population level. By comparing the average biological aging rate at the population level with the biological state of individuals, we aim to measure the relative speed of an individual’s aging compared to the population average. (3) We will compare the aging models constructed from various databases to identify differences in human aging clocks between different populations, such as European and Asian groups.

Scientific Rationale: Although the intrinsic logic behind the real causes of aging is not yet fully understood, the actual aging process manifests externally. These external manifestations, which may include complex changes in epigenetics, proteins, and metabolites, can potentially provide insights into the true biological aging process. Statistical methods, such as machine learning, offer the possibility to decipher these complex changes. Therefore, we plan to employ machine learning statistical methods to elucidate the relationship between the complex changes in epigenetics, proteins, and metabolites and chronological age.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/construction-of-a-multidimensional-analysis-model-for-the-onset-progression-treatment-and-prognosis-evaluation-of-cardio-cerebrovascular-diseases

Construction of a multidimensional analysis model for the onset, progression, treatment, and prognosis evaluation of cardio-cerebrovascular diseases

Last updated:
ID:
242487
Start date:
10 March 2025
Project status:
Current
Principal investigator:
Professor Xuegong Zhang
Lead institution:
Tsinghua University, China

Aims: We aim to integrate and analyze multi-omics data, including epidemiology, imaging, genomics, and other relevant factors, to construct a multidimensional analysis model for cardio-cerebrovascular diseases. This model will facilitate the identification of high-risk populations and assist in making precise treatment strategies.
Scientific rationale: Cardio-cerebrovascular diseases represent significant global contributors to mortality and disability, impacting a broad demographic spectrum with a high prevalence. Variability in the occurrence, progression, treatment response, and prognosis of these diseases among individuals is markedly influenced by diverse factors such as lifestyle, age, environment, genetic elements, and genetic polymorphisms. Consequently, the early diagnosis and long-term management of cardio-cerebrovascular diseases hinge upon the theoretical underpinnings and clinical expertise of physicians. The flexible adjustment of suitable treatment plans based on individual characteristics of different patients highly depends on the accumulated experience of physicians over time. To address the experiential diversity among physicians, this study aims to comprehensively integrate multidimensional data from cardio-cerebrovascular diseases cohorts, encompassing epidemiological, demographic, environmental, psychological, lifestyle, laboratory findings, electrocardiographic patterns, cardio-cerebrovascular imaging traits, and genomic profiles. A multidimensional risk model for cardio-cerebrovascular diseases will be developed, facilitating phenotype differentiation analysis, disease risk assessment, treatment response prediction, and long-term prognostic evaluation based on multidimensional datasets.
Project duration: The project is expected to span approximately 3 years, during which the associations between data from different dimensions and cardio-cerebrovascular diseases will be analyzed individually and subsequently amalgamated into the multidimensional analytical framework.
Public health impact: By constructing a multidimensional analysis model, we plan to use necessary demographic information and medical examination data to assist physicians in the early identification of high-risk population for cardio-cerebrovascular diseases. The model will also predict the potential differences in treatment response to different treatment options, helping physicians develop more individualized and precise treatment plans. In addition to medical practice, the model will further reveal the significant phenotypic variability within the vast patient population of cardio-cerebrovascular diseases through the analysis of multi-omics data from multiple dimensions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/construction-of-a-normative-database-of-human-retinal-thickness-from-existing-biobank-participants

Construction of a Normative Database of Human Retinal Thickness from Existing BioBank Participants

Last updated:
ID:
61229
Start date:
21 September 2021
Project status:
Current
Principal investigator:
Mr Aaron Carass
Lead institution:
Johns Hopkins University, United States of America

Imaging of the human eyes provides a unique insight into the health of the body. Measuring the thicknesses of the retinal layers is beginning to serve as an important and readily accessible biomarker. The changing thicknesses of the retinal layers over the course of normal human aging have been shown to be associated with changes in the brain. These changes have been associated with numerous neurodegenerative diseases, including multiple sclerosis, Alzheimer’s disease, Parkinson’s disease, and others. Therefore it is important to we provide detailed measurements of the human retina across the BioBank participants. In particular, highlighting the individual differences in the changing human retina and their correlations with the brain ageing process. We hope, that by providing detailed measurements of the human retina across the BioBank participants, we can identify at risk patients and schedule secondary screening earlier.

Our primary goal is to provide a public database of the thickness of the human retina as well as the thicknesses of several of the tissue layers within the retina. This will cover the full age range of available BioBank participants. This database will cover the
subdivisions typically used by ophthalmologist in the retina; known as the ETDRS grid, which subdivides the retina into nine regions of interest. Thus the database will provide location specific measures of normal thickness for the various retinal layers. A secondary goal, will be exploring the potential of the ETDRS retinal thicknesses in identifying neurodegenerative diseases earlier. This will be based on analysis of the retinal thicknesses as biomarkers for disease that will involve trying to formulate these biomarkers into a generalized disease progression score; similar to the Alzheimer’s disease progression score (ADPS). The ADPS is an approach for exploring the time-dependent changes of biomarkers related to Alzheimer’s disease. The key idea being that different biomarkers change at different times and rates. The change of no single biomarker identifies the onset of disease but the collection biomarkers may offer insight into the neurological health of a patient. Our initial study is anticipated to take two years; with our database of retinal thicknesses being updated as the BioBank continues to enroll participants.

With our study, we hope to identify those who may be at risk of neurodegeneration. Thus allowing disease modifying therapies to be introduced at any earlier stage–improving patient prognoses.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/construction-of-a-reversal-model-for-mild-cognitive-impairment

Construction of a reversal model for mild cognitive impairment

Last updated:
ID:
144839
Start date:
22 November 2023
Project status:
Current
Principal investigator:
Dr Xinyi Xu
Lead institution:
Hebei Medical University, China

Aims!This study aims to investigate the stability and trajectory of cognitve function in older adults as well as to explore the lifestyle factors (e.g. leisure activities, diet, and sleep) and biochemical markers (e.g. inflammatory index, blood glucose, blood lipid level) associated with reversion from mild cognitive impairment (MCI) to normal cognitive function (NC).
Scientific rationale: MCI is considered to be an intermediate state between normal cognitive aging and early dementia, and it is a premorbid risk factor for dementia. However, the natural progression of MCI is not always linear. For example, previous MCI conversion studies reported that considerable numbers of MCI cases can either be reversible or remain stable over a long period of time. Studies have shown that nearly 24% of people with MCI can finally revert to normal cognitive function (NC). Most studies have focused on the progression from MCI to dementia, while relatively little attention has been paid to the reversion from MCI to NC.
Identifying the underlying mechanism or predictive factor for the cognitive recovery is crucial because it would provide prognostic values for dementia-related cognitive decline, as well as help with developing an intervention program. In addition, because of the existence of four MCI subtypes (mnestic MCI-Single Domain, Amnestic MCI-Multiple Domain, Non-Amnestic MCI Single Domain, and Non-Amnestic MCI-Multiple Domain), the literature on the meaning of reversion from MCI to CN remains inconclusive. Therefore, understanding the meaning of reversion from MCI to CN is important for both research and clinical practice.
Project duration: This project will last about three years, including data cleaning, imputation of missing value, data recoding, and data analysis.
Public health impact: At present, dementia is a global disease, and every three seconds occurs one new case of dementia globally. Dementia has become one of the important public health problems, and it brings a huge burden on patients, their families and the whole society. Therefore, it is critical to identify potential mechanisms and predictors of cognitive recovery in people with MCI, which will provide an important basis for preventing dementia-related cognitive decline.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/construction-of-chronic-disease-risk-factors-and-chronic-disease-time-series-co-morbidity-network

Construction of chronic disease risk factors and chronic disease time-series co-morbidity network

Last updated:
ID:
570161
Start date:
25 February 2025
Project status:
Current
Principal investigator:
Dr Yu-zhe Kong
Lead institution:
Second Xiangya Hospital of Central South University, China

The global rise in chronic diseases like cardiovascular diseases, metabolic disease and chronic respiratory conditions demands an exploration of their multifaceted causes-genetic, environmental, lifestyle, and clinical. However, few studies have examined the risk factors for chronic diseases and the sequence of diseases caused by chronic diseases in terms of their temporal order of prevalence.

This project aims to explore the intricate causes and progression of chronic diseases by building networks that map their risk factors and co-morbidities, utilizing advanced statistical and machine learning methods. This research will provide vital insights for developing targeted prevention and treatment strategies.

We’ll use advanced statistical techniques such as generalized estimating equations, Cox proportional hazards models, logistic regression, and restricted cubic splines to model nonlinear relationships. Additionally, transcriptional, protein, metabolic, and epigenetic multi-omics revalidation was also performed using machine learning, survival, network, GWAS, temporal sequencing, and clustering pathway enrichment analyses.

Over the next three years, our program will focus on two research questions: what are the risk factors for chronic diseases and what is the sequence in which chronic diseases occur (i.e., constructing time-series co-morbidity networks for chronic diseases).

We will focus on Chronic Kidney Disease, Circulatory Disease (cardiovascular disease), Mental Disease (Depression, Anxiety, etc.), Motor System Disease (Osteoarthritis, Sarcopenia), Digestive Disease (NAFLD), etc.. We will construct a network of chronic disease co-morbidities as well as risk factors for chronic diseases from four major aspects: genes, lifestyle habits, environment, and biomarkers.

The results of the research will be disseminated in the form of publications.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/construction-of-multi-dimensional-statistical-shape-model-for-a-heart

Construction of Multi-dimensional Statistical Shape Model for a heart

Last updated:
ID:
42239
Start date:
31 August 2018
Project status:
Current
Principal investigator:
Dr Shoko Miyauchi
Lead institution:
Kyushu University, Japan

Statistical Shape Model (SSM) of a target organ quantitatively describes the average and variance of the organ among individuals. Since the SSM enables to estimate the whole shape of a target organ from its partial shape data, SSM is used in various computer-aided treatment and diagnosis systems. Conventional SSMs describe the shape variation of motionless organs (e.g. bone). On the other hand, few description methods consider the deformation of organs (e.g. heart). The aim of our research is to construct a new multi-dimensional SSM for the heart that describes not only inter-individual differences of cardiac shape but also the cardiac deformation. One potential application using the multi-dimensional SSM for the heart is to estimate the movement of the patient heart in a cardiac cycle from the partial shape of the patient heart at a certain time. Accordingly incorporating the multi-dimensional SSM for the heart into a computer-aided system leads to the improvement of the efficient and highly accurate treatment / diagnosis for hearts.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/construction-of-multimodal-symbiosis-medical-database

Construction of multimodal symbiosis medical database

Last updated:
ID:
88365
Start date:
16 November 2022
Project status:
Current
Principal investigator:
Dr Huiying Liang
Lead institution:
Guangdong Provincial People's Hospital, China

The UK Biobank project is a large prospective cohort study, and it tracks the health of~ 500,000 individuals from across the United Kingdom. Multimodal data including health records, medical image and genetics have been collected. The correlation and complementarity between multimodal data make it possible to describe the characteristics of the disease more comprehensively at the same time. However, there are some problems such as modality incompleteness and modal weights imbalance in the process of multimodal medical data analysis. We aim to build new algorithm frameworks to impute missing data, and to predict the overall disease status and survival of participants with obesity, dyslipidemia, hyperglycemia, or hypertension at the baseline visit. Our work will last for 36 months and help us to more accurately excavate the complex characteristics of diseases, and to support for subsequent intelligent decisions and predictions more powerful.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/contextual-data-analytics-for-clinical-decision-making

Contextual data analytics for clinical decision making

Last updated:
ID:
49398
Start date:
1 August 2019
Project status:
Closed
Principal investigator:
Dr Matthias Siebert
Lead institution:
Siemens HealthCare GmbH, Germany

Due to recent breakthroughs in molecular diagnostics and imaging, the amount of clinically relevant data is constantly growing. As a result, medicine and healthcare are drifting towards being ‘big data’ disciplines. Precision medicine refers to the idea of delivering the right treatment to the right patient at the right time. In order to enable precision medicine and to improve the quality of healthcare in the future, we need methods that integrate complex and heterogeneous clinical data and translate it into actionable clinical knowledge.
This study aims to tackle both requirements. We start with bringing the different types of data provided by the UK Biobank in context with each other and with existing biological and clinical knowledge from public databases. For instance, to assess the association of genetic variants with a specific clinical outcome, single nucleotide variants will be considered both in their biological context – e.g. affected genes, biological pathways and known implications for health and disease – as well as their clinical context – e.g. other physical measures and patient socio-demographics and lifestyle.
This will allow us to gain a more holistic view on specific clinical pictures and to then develop machine learning algorithms that are particularly suited to leverage this contextual information and the rich relationships between single data entities for supporting clinical decision making – e.g. assessing the risk of developing a certain disease or discovering new disease endotypes.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/contribution-of-dna-variants-to-the-risk-of-type-2-diabetes-and-associated-disorders

Contribution of DNA variants to the risk of type 2 diabetes and associated disorders

Last updated:
ID:
702482
Start date:
16 June 2025
Project status:
Current
Principal investigator:
Dr Amelie Bonnefond
Lead institution:
INSERM, France

In this proposal, we seek to validate associations between loss-of-function and gain-of-function DNA variants and the risk of metabolic disorders (or related trait levels) using the UK Biobank, building on findings obtained from French participants. These variants will be located either (i) within coding exons of candidate genes implicated in metabolic disorders such as type 2 diabetes, obesity, kidney diseases, and cardiovascular diseases, or in associated traits, or (ii) in non-coding regulatory regions, such as active enhancers or promoters in human metabolic tissues. To evaluate the functional impact of these variants, we will employ in silico and/or in vitro approaches. This project aims to uncover novel functional genetic markers and pathways involved in metabolic disorders, ultimately contributing to population stratification for precision medicine.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/contribution-of-energy-protein-intake-and-the-interaction-with-physical-activity-to-transitions-between-frailty-states-prospective-analysis-of-the-uk-biobank

Contribution of energy, protein intake and the interaction with physical activity to transitions between frailty states: prospective analysis of the UK Biobank

Last updated:
ID:
103200
Start date:
10 October 2023
Project status:
Current
Principal investigator:
Dr Nuno Mendonça
Lead institution:
University of Évora, Portugal

The European population is ageing but, for many countries, the extra years of life are not free of disability. Compressing morbidity into the later years of life is of special interest; not only to increase quality of life, but also to relieve the immense strain on the European healthcare systems. Frailty is a clinical syndrome defined as an increased vulnerability or failure to return to a healthy equilibrium after a stressor event that increases the risk of dependency, care home admission, hospitalisation, and death. Frailty is defined by five criteria: muscle weakness, slow walking, low activity, exhaustion, and unintentional weight loss. Diet and physical activity are major modifiable risk factors for morbidity, disability, and death and are central do these five frailty criteria. Having a healthy diet with enough energy and protein coupled with physical activity may slow down the appearance of frailty above and beyond what each would be able to achieve by itself. Therefore, for the next 2 years we will use the UK Biobank to try to determine the contribution of energy intake, protein intake and physical activity to the transitions (progression or recovery) to and from frailty states (robust, pre-frail and frail) in individuals aged 50 and over. The results will inform dietary guidelines for older adults as well as their relationship with different levels of physical activity, and possibly change the approach to combat frailty in Europe.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/contribution-of-rare-dna-variants-to-the-risk-of-metabolic-disorders-and-to-the-level-of-associated-metabolic-traits

Contribution of rare DNA variants to the risk of metabolic disorders and to the level of associated metabolic traits

Last updated:
ID:
67575
Start date:
27 January 2021
Project status:
Current
Principal investigator:
Dr Amelie Bonnefond
Lead institution:
INSERM, France

Aims: In this proposal, based on our data in a French population, we aim to confirm in the UK Biobank association signals between specific DNA variants and genetic regions, and the risk of various metabolic disorders.
Scientific rationale: Diabetes affects 420 million patients worldwide and that number will increase to 700 million by 2030. Diabetes is the sixth leading cause of mortality. Type 2 diabetes (T2D) represents more than 90% of all diabetes cases. Diabetes is a complex genetic disorder, with 72% of heritability. The worldwide prevalence of obesity nearly tripled between 1975 and now. Such as T2D, obesity is highly heritable (~70% heritability). Two billion people are currently overweight, and their co-morbidities represent a major medical burden and challenge of the health care system. Obesity and T2D are leading causes of kidney diseases, which happens in many patients with T2D and/or obesity. Many of these patients will progress to end-stage renal disease requiring dialysis. Large international studies including those using the UK Biobank have identified one thousand DNA regions associated with T2D risk, obesity, kidney diseases, or associated with lipid traits. However, they still only explain ~20% of disease heritability. In 2012, we demonstrated the contribution to T2D risk of rare deleterious mutations affecting the melatonin receptor (melatonin being a key controller of the day/night clock). Since then, many studies have highlighted the contribution to common disease of rare mutations impairing important protein of metabolism. We have recently sequenced the genome of 10,000 French individuals with T2D, obesity, kidney diseases and normal controls and identified many mutations of potential interest. We would like to use the UK Biobank to verify and expand our data.
Project duration: 5 years
Public health impact: This project should lead to the identification of new genes and mechanisms involved in the development of metabolic disorders as the first stage towards the establishment of the 21st century 4P Precision Medicine that should be Predictive, Preventive, Personalized and Participatory.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/control-data-sets-for-congenital-heart-disease-copy-number-variant-analysis

Control data-sets for congenital heart disease copy number variant analysis

Last updated:
ID:
19348
Start date:
11 May 2016
Project status:
Closed
Principal investigator:
Professor Peter Gruber
Lead institution:
University of Iowa, United States of America

We are studying the genetic basis of congenital heart disease. Specifically we are interested in genetic rearrangements that may occur in these children and adults. We are interested in improving the health of children and adults with congenital heart disease. It has been clearly show that children with identified genetic abnormalities have an increased risk of morbidity and mortality compared to those without obvious identifiable genetic abnormalities. However, no study to date has comprehensively looked at CNV (copy number variants) in this population. We are completing this study but need a control population in order to compare. We will use the data from the UK Biobank to compare to our other data. This comparison will allow us to determine which genetic abnormalities are distinct for the population with congenital heart disease compared to that without. 4,000 normal subjects without known congenital heart disease


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/controllable-synthesis-of-virtual-heart-populations-for-in-silico-trials-of-medical-devices

Controllable synthesis of virtual heart populations for in silico trials of medical devices

Last updated:
ID:
392951
Start date:
9 April 2025
Project status:
Current
Principal investigator:
Dr Nishant Ravikumar
Lead institution:
Adsilico Limited, Great Britain

Despite rigorous testing requirements for medical device approval, around 83,000 patient deaths and injuries have been linked to implanted devices over the past decade. Conventional human trials struggle to recruit sufficiently diverse patients, while animal testing raises ethical concerns and may not accurately represent the target patient population. This project aims to develop and validate advanced computational techniques to synthesise large virtual populations of 3D patient anatomies that are statistically representative of real-world diversity. Unlike current methods limited by patient data availability, our unique generative modelling approach can combine information across multiple datasets to create plausible but fully synthetic anatomies. Importantly, the synthesis process can be controlled to match target patient demographics, enabling more inclusive device evaluation. The project will focus on generating diverse virtual heart models to support simulation-based (in silico) testing of a variety of cardiovascular devices. Success will lead to more affordable, ethical and representative device testing to ensure patient safety while reducing costs and time to market for new medical technologies. Validated synthetic datasets have potential for broader commercialisation to medical companies undertaking regulatory submissions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/controlling-structure-induced-variations-in-non-invasive-perfusion-mri-of-neurodegeneration

Controlling Structure Induced Variations in Non-Invasive Perfusion MRI of Neurodegeneration

Last updated:
ID:
43172
Start date:
4 November 2019
Project status:
Closed
Principal investigator:
Professor Michael Chappell
Lead institution:
University of Oxford, Great Britain

Perfusion imaging allows us to measure the vital role played by delivery of blood to the brain in keeping it supplied with nutrients and removal of waste. Any deviations of the blood supply from normal can be a sign of disease. In particular early and subtle changes in perfusion might mark regions of the brain which are affected by degenerative diseases such as dementia before other imaging signs become obvious.

The technology exists and is increasingly widely available to image perfusion quickly and safely using Magnetic Resonance Imaging. Thus perfusion Magnetic Resonance Imaging could be a valuable tool in the understanding of dementias, as well as the diagnosis and monitoring of patients with dementia. The challenge that remains is making sufficiently specific measurements of subtle changes in blood supply that would be needed to make the technology truly useful for patients. This project addresses that problem in three ways:

> Automated removal of errors associated with imperfect measurement, for example due to motion of the patient.

> Methods to control for differences between patients due to their individual brain structure, allowing blood supply measurements to be compared between individuals or from a patient to a population of similar healthy adults. These methods remove uncertainties introduced by other differences between the brain’s of individuals that are not related to perfusion.

> Generation of personalised reference perfusion images for an individual patient against which their measured perfusion can be compared to detect changes specific to that individual.

The methods and tools that are to be generated in this project will enable perfusion Magnetic Resonance Imaging to be used more effectively in the UK-wide effort to understand dementia and in the search for new and effective treatments. Ultimately the work done in this project will enable perfusion Magnetic Resonance Imaging to become a valuable clinical tool that can be used in the diagnosis and monitoring of individual patients with dementia.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/convergent-and-divergent-genetic-and-epigenetic-mechanisms-in-neurodegenerative-disorders

Convergent and Divergent Genetic and Epigenetic Mechanisms in Neurodegenerative Disorders

Last updated:
ID:
1009383
Start date:
24 September 2025
Project status:
Current
Principal investigator:
Professor Eran Meshorer
Lead institution:
Hebrew University of Jerusalem, Israel

This project aims to investigate the genetic and epigenetic mechanisms underlying neurodegenerative disorders, with a particular focus on polyglutamine (PolyQ) diseases such as Huntington’s disease and Spinocerebellar ataxias, as well as Amyotrophic Lateral Sclerosis (ALS). Although these disorders are clinically distinct, they may share overlapping pathways or diverging molecular processes that contribute to neurodegeneration. Understanding these patterns is critical for improving disease classification and identifying potential therapeutic targets (Li Gan et al., 2018).

The study will primarily use UK Biobank’s whole genome sequencing (WGS) and methylation array data to explore the convergence and divergence of genetic risk factors across these diseases. We will perform genotype-first analyses of known or suspected pathogenic variants in key neurodegenerative genes (such as HTT, ATXN1, C9orf72), and examine the downstream phenotypic and clinical profiles of variant carriers compared to non-carriers. These analyses will include assessments of disease status, cognitive function, and age-related traits.

A second key component of this project involves integrating findings from external single-nucleus RNA sequencing (snRNA-seq) studies to guide the selection of candidate genes and regions of interest. We aim to conduct both genome-wide and targeted analyses, including rare variant burden testing, polygenic risk scoring, and sequence alignment at loci associated with trinucleotide repeat expansions.

All analyses will be conducted through the Research Analysis Platform (RAP). The project is led by graduate student Rivka Masar as part of her academic research, under the supervision of the Principal Investigator, who will ensure compliance with UK Biobank requirements. We request access to sequencing, methylation, clinical, cognitive, and mortality data, and seek approval under the student fee structure.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/copd-follow-up-access-to-genotype-and-phenotype-information-and-dna-samples

COPD follow up ? access to genotype and phenotype information and DNA samples

Last updated:
ID:
24299
Start date:
1 January 2017
Project status:
Current
Principal investigator:
Mr Frosti Jonsson
Lead institution:
deCODE Genetics, Iceland

deCODE’s ongoing genetic study includes 6,000 COPD cases and 300,000 controls. Many sequence variants reported to associate with COPD or lung function replicate in our dataset. We have many novel variants associating with COPD, requiring follow up in a large dataset like the UKBiLEVE cohort, in particular rare variants identified by whole genome sequencing of the Icelanders. Our aim is to use existing genotypes or where necessary to undertake further genotyping of samples from UKBiLEVE subjects to confirm associations found in the Icelandic population, thereby establishing novel, robust associations between sequence variants and COPD or lung function. Identifying novel variants in the genome that associate with COPD or lung function will contribute to increased understanding of the disease and may provide new targets for development of improved treatments for the disease. Existing genotypes for UKBiLEVE subjects will be analysed for selected variants to confirm novel associations with COPD or its subphenotypes, emphysema and chronic bronchitis, or quantitative lung function traits, already identified in the deCODE sample set. DNA from UKBiLEVE subjects will be genotyped for rare variants for which no genotypes exist in UK Biobank. To identify UKBiLEVE subjects with COPD, emphysema, chronic bronchitis or asthma, information on self-reported or HES diagnosis of those diseases would be needed. Genotype information for 48931 individuals from UKBiLEVE and DNA samples from a subset of 19017 individuals, defined as COPD cases or controls in the study, are requested. In addition, we request genotype information and DNA samples for with self reported, or registered diagnosis of, COPD, emphysema or chronic bronchitis for up to 7428 individuals, fewer if the selected groups overlap. In total, genotypes for 56359 and DNA for 26445 individuals are requested.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/copy-number-variants-in-cardiovascular-and-neuropsychiatric-disorders

Copy-number variants in Cardiovascular and Neuropsychiatric disorders

Last updated:
ID:
19056
Start date:
1 November 2016
Project status:
Closed
Principal investigator:
Dr Simon Williams
Lead institution:
University of Manchester, Great Britain

Rare genomic copy-number variants (CNVs) have been implicated as causative factors in a number of complex diseases including schizophrenia, autism and congenital heart disease (CHD). Many factors are known to increase the risk of these diseases and, in recent years, developments in sequencing and array-based platforms have enabled investigation into the genetic factors that contribute to these conditions. Previous studies have identified rare CNVs in specific regions that associate with these diseases but there has been relatively little investigation into the occurrence of these CNVs in the wider population. Using the data generated through the UK Biobank?s genotyping project, the aim of this study is to assess the prevalence of rare CNVs in the UK population. Improved characterisation of rare CNVs associated with disease will facilitate gene discovery and in turn develop increased understanding of the influence of genetics in complex disease. This will ultimately impact future healthcare as it will enable earlier diagnosis in children and families affected by these conditions, improving the development of personalised care pathways. Using the Biobank genotyping data, we will call CNVs and compare these with CNVs previously implicated in disease phenotypes to gain a greater insight into the prevalence of these ‘rare’ events in the wider population. This study would use the UK Biobank genotyping data (full cohort).


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/copy-number-variation-from-whole-exome-sequencing-in-the-uk-biobank

Copy number variation from whole exome sequencing in the UK Biobank

Last updated:
ID:
49978
Start date:
10 October 2019
Project status:
Current
Principal investigator:
Dr Tomas Fitzgerald
Lead institution:
European Bioinformatics Institute (EBI), Great Britain

Copy number variation (CNV) is an important source of genetic differences between individuals and can give raise to observable traits. Certain CNVs are known to cause extreme traits in human and can be the direct cause of a number of severe illnesses. In this project we aim to make use of next generation sequencing datasets across a large UK cohort, to create detailed CNV maps, and to perform robust association testing of copy number variable regions against human phenotypic measurements with a particular focus on brain structure and function.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/core-business-of-the-cnv-analysis-group-for-the-psychiatric-genomics-consortium

Core business of the CNV analysis group for the Psychiatric Genomics Consortium

Last updated:
ID:
62713
Start date:
13 July 2020
Project status:
Current
Principal investigator:
Professor Jonathan Sebat
Lead institution:
University of California, San Diego, United States of America

This project will utilize genetic data from the UK Biobank to carry out large scale studies of copy number variation (CNV) in psychiatric disorders. This project will be carried out by the CNV analysis group for the Psychiatric Genomics Consortium. CNV calls will be generated across the full UKBB dataset, and the UKBB data will be combined with existing data from 11 PGC disorders including Autism, ADHD, Bipolar Disorder, Schizophrenia, Major Depression, Tourettes/OCD, PTSD, Substance Use Disorders, Eating disorders, and Alzheimers disease, thereby significantly increasing the numbers of population controls (and cases if diagnosis information is available). The association of CNVs with psychiatric diagnosis will be examined, and the influence of CNVs on cognitive, physiological and anthropometric traits in the general population will be examined.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/correcting-finescale-population-stratification-using-haplotype-sharing-to-improve-association-study-and-polygenic-risk-score-accuracy

Correcting finescale population stratification using haplotype sharing to improve association study and polygenic risk score accuracy

Last updated:
ID:
94132
Start date:
6 December 2022
Project status:
Closed
Principal investigator:
Dr Ross Patrick Byrne
Lead institution:
Trinity College Dublin, Ireland

Any given two humans will share at least one ancestor at some point in the past. This shared ancestry may be reflected in their genomes as shared stretches of DNA. The number of these ancestral segments shared between individuals relates to the degree of shared ancestry between them. A recent study in Britain (“The People of the British Isles”) which examined the patterns of ancestral segment sharing across the Island identified subtly related genetic subgroups of people that correspond to geographic regions in Britain. For example individuals sampled from Devon form a subtly distinct subgroup from individuals sampled from Cornwall. Studies in Ireland, Finland, Japan, Italy, The Netherlands, France and Spain have since revealed subtle genetic subgroups within these countries based on patterns of ancestral segment sharing. The existence of these ancestrally enriched subgroups has important implications for the design of genetic association studies performed on the UK BioBank and other datasets, as individuals within these clusters are expected to share slightly more genetic variation than random due to their shared ancestry. As such if we are looking for a mutation which shows association with an outcome such as a disease without accounting for this ancestral similarity we may falsely identify one simply shared due to ancestry.

Current methods for detecting and correcting underlying shared ancestry in association studies show lower resolution in detecting subtle shared ancestry than methods using ancestral segments, and have failed to identify within country subgroups. Hence it is possible that applying new methods leveraging segment sharing in the context of correcting for shared ancestry association studies will make results more robust and reduce false associations.

Our study aims to explore the use of a fast and scalable method for looking at shared ancestry segments across individuals in the UK Biobank. We will compare the outcomes of using this method to correct association studies to those from the standard methods and investigate the degree of inflation due to population structure in each using an established method called LD-score regression. Our project should take between 1 and 2 years and will provide a reusable resource for all Biobank researchers. Ideally, if successful, our project should also reduce the rate of false positive associations, allowing us to have greater confidence in results from the Biobank, and better inform targets for drug development and potentially improve the accuracy of genetic prediction, which may have clinical applications in the future.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/correlates-and-consequences-of-cardiometabolic-multimorbidity

Correlates and Consequences of Cardiometabolic multimorbidity

Last updated:
ID:
13784
Start date:
1 July 2015
Project status:
Current
Principal investigator:
Professor Emanuele Di Angelantonio
Lead institution:
University of Cambridge, Great Britain

As many cardiometabolic diseases (such as myocardial infarction, stroke, hypertension and diabetes mellitus) share risk factors, these conditions often co-occur (?cardiometabolic multimorbidity?). However, despite increases in the prevalence of such multimorbidity, there is a paucity of evidence concerning the potential consequences for survival. This application proposes to:

1) assess the prevalence of cardiometabolic multimorbidity in a contemporary population

2) identify potential genetic and non-genetic determinants of cardiometabolic multimorbidity

2) estimates the associations of cardiometabolic multimorbidity with the risk of subsequent major health events and mortality. This research is in the public interest considering population aging and that people who have cardiometabolic multimorbidity has been increasing rapidly worldwide. Evidence concerning the potential consequences of cardiometabolic multimorbidity for survival could importantly inform public health priorities and the targeting of prevention efforts.
We will perform analyses investigating the prevalence of cardiometabolic diseases (such as myocardial infarction, stroke, hypertension and diabetes mellitus) in isolation and in combination. We will assess the associations of cardiometabolic multimorbidity with other prevalent diseases as well as with socio-demographic, lifestyle, environment, early life, psychosocial and physical measures. When available in UK Biobank, we will analyse the genetic data to assess possible genetic determinants of cardiometabolic multimorbidity.
Once suitable numbers of disease outcomes have accrued we will examine the relationships of cardiometabolic multimorbidity with future health outcomes.
The full cohort data will be required.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/correlates-of-an-anorexia-nervosa-polygenic-resilience-score-in-a-general-population-cohort

Correlates of an Anorexia Nervosa-polygenic resilience score in a general population cohort

Last updated:
ID:
827551
Start date:
19 August 2025
Project status:
Current
Principal investigator:
Professor Nadia Micali
Lead institution:
Region Hovedstadens Psykiatri, Denmark

Scientific rationale:
Anorexia nervosa (AN) is a complex disorder influenced by genetic, environmental, and neurobiological factors. Polygenic risk scores (PRS) have been identified for AN susceptibility, but the concept of genetic resilience, variants that protect against disease, remains understudied. Some individuals with high genetic risk do not develop AN, potentially due to resilience alleles that buffer risk loci (Hess et al., 2021; PMID: 31492941).

Research Questions:
1. Do genetic factors confer resistance to AN despite high genetic risk?
2. What is the heritability of AN resilience, and does it share causal variants with AN and other traits?
3. How does the AN resilience PRS correlate with psychological, metabol-ic, and anthropometric traits?

Aims:
1. Calculate an AN PRS in two cohorts (UK Biobank, Danish cohort).
2. Identify resilient vs. non-resilient individuals and conduct an AN resilience GWAS meta-analysis.
3. Derive an AN resilience PRS
4. Explore heritability for resilience to AN and perform colocalization anal-ysis to test for shared genetic causal variants with AN and other traits.
5. Explore genetic correlations and associations of the AN resilience PRS with diverse phenotypes.

Methods:
We will construct an AN PRS in the UK Biobank and Danish cohort. AN cases and controls with high AN PRS will be considered as “high-risk subsample”. This sub-sample will be determined by using in the 90th percentile and above of the AN PRS in the controls as lower and upper bound, respectively. AN cases within this range will be considered as non-resilient, while controls without AN in the same range will be considered as resilient. A GWAS of AN resilience will be conducted and meta-analyzed across cohorts.

The resilience GWAS will be used to calculate an AN resilience PRS in two addi-tional cohorts, assessing its correlation with diverse traits. Additional analyses in-clude fine-mapping, SNP-based heritability and colocalization analyses.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/correlates-of-health-related-variables-quality-of-life-physical-function-and-cancer-related-outcomes-among-people-with-or-without-a-cancer-diagnosis

Correlates of health-related variables, quality of life, physical function, and cancer-related outcomes among people with or without a cancer diagnosis.

Last updated:
ID:
92482
Start date:
16 November 2022
Project status:
Current
Principal investigator:
Dr Cynthia Forbes
Lead institution:
University of Hull, Great Britain

The number of people living with and beyond cancer is growing. Research shows we could have fewer cancer cases, cancer-related deaths, and healthcare costs if people were more physically active and adopted a healthier lifestyle. Regular physical activity, together with healthy eating, helps people maintain their daily activities and improves their energy levels and quality of life. However, many older adults living with or beyond cancer are inactive with an unhealthy diet. More research into how lifestyle behaviours can help improve physical, emotional, and mental health and in turn cancer recovery is urgently needed. This is also the case for people who are about to receive surgery or treatment for their cancer diagnosis.
We want to look at the data and see if there are any relationships or patterns among people with better health outcomes and if there is any difference between those with a history of cancer and those without. This will take the form of a number of different statistical tests that will look for relationships in demographic characteristics, clinical and physical measures, and information about lifestyle behaviours. These relationships are important to find and understand because it can help us predict who might need what specific support in order to live as well as possible, as long as possible after a cancer diagnosis.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/correlating-genotype-with-phenotype-for-variants-in-rare-disease-genes-in-the-uk-biobank

Correlating Genotype with Phenotype for Variants in Rare Disease Genes in the UK Biobank

Last updated:
ID:
66057
Start date:
8 March 2021
Project status:
Current
Principal investigator:
Professor Anna Michelle Lehman
Lead institution:
University of British Columbia, Canada

Rare diseases affect at least 5% of the population, which means cumulatively, they are a major health concern. More than 70% of rare diseases are caused by genetic variants. Despite being born with a potentially disease-causing variant, a person may not develop symptoms until later in life, if at all. If the symptoms are not severe and specific enough, a genetic diagnosis may not be sought or made. Our research seeks evidence of milder signs of certain rare genetic diseases in those who may carry a weaker variant than that which usually causes the full rare disease. In another aim, we seek evidence whether or not variants previously classified as disease-causing are associated with bad outcomes, specifically in genes being considered for intentional screening in a general population. Finally, our research program seeks novel causes for rare genetic disorders, and the UK Biobank data provide valuable comparison data.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/correlation-analysis-between-metabolism-related-risk-factors-and-long-term-prognosis-of-metabolic-syndrome

Correlation analysis between metabolism-related risk factors and long-term prognosis of metabolic syndrome

Last updated:
ID:
888618
Start date:
19 September 2025
Project status:
Current
Principal investigator:
Dr Hong Zhu
Lead institution:
The First Affiliated Hospital of Wenzhou Medical University., China

Metabolic syndrome refers to a cluster of metabolic disorders that significantly increase the risk of developing cardio-cerebrovascular diseases, type 2 diabetes, and other health complications. While traditional factors such as age, ethnicity and lifestyle are established contributors to metabolic syndrome onset and progression , the roles of genetic susceptibility, multi-omics biomarkers, imaging-derived phenotypes, and environmental exposures in shaping long-term metabolic syndrome prognosis remain inadequately characterized.
To quantify how psychological, cognitive, genetic, metabolic, imaging and environmental factors shape long-term metabolic syndrome outcomes.
In this study, datasets from the UK Biobank were utilized, including baseline questionnaires (e.g., lifestyle, cognitive function, and environmental exposures), genotyping data, physical measurements, metabolic biomarkers, longitudinal health records, clinical outcomes (e.g., macrovascular and microvascular complications, cancer, mental disorders), and imaging data. Using statistical methods such as the Cox proportional hazards model, logistic regression model, mediation analysis, restricted cubic splines, we evaluated the independent effects, dose-response relationships, and interactions of relevant risk factors. Furthermore, by integrating data sources, a analysis was conducted to investigate the associations between risk factors (e.g., lipid profiles, dynamic changes in blood glucose, obesity indicators, genetic susceptibility, environmental exposure, and mental and cognitive characteristics) and the long-term prognosis of metabolic syndrome, including all-cause mortality, cardiovascular and cerebrovascular events, diabetic complications, and cancer incidence.
This study focuses on the interplay mechanisms of genetics, metabolism and environment, to provide a scientific basis for clinical decision-making and novel drug researches on metabolic syndrome.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/correlation-and-causal-relationship-of-blood-lipids-and-metabolites-healthy-lifestyle-and-socioeconomic-status-with-prevalence-and-mortality-of-metabolic-disease

Correlation and causal relationship of Blood lipids and metabolites, healthy lifestyle and socioeconomic status with prevalence and mortality of metabolic disease.

Last updated:
ID:
479734
Start date:
18 October 2024
Project status:
Current
Principal investigator:
Professor Yun Zhang
Lead institution:
Peking Union Medical College Hospital (PUMCH), China

Metabolic diseases, such as diabetes, cardiovascular disease, gout, and metabolic syndrome, represent a significant global health burden. The interplay between blood lipids, metabolites, lifestyle factors, and socioeconomic status (SES) is complex and multifactorial. Elevated blood lipids and altered metabolites are well-established risk factors for metabolic diseases, yet their interaction with lifestyle behaviors (e.g., diet, physical activity, smoking) and SES remains inadequately explored. Understanding these relationships is critical for developing targeted interventions to reduce the incidence and mortality of metabolic diseases.
Objectives:
1. To evaluate the correlation between blood lipids, metabolites, and metabolic disease prevalence and mortality.
2. To assess the impact of healthy lifestyle factors (e.g., diet, physical activity, smoking cessation) on the relationship between blood lipids and metabolic disease outcomes.
3. To investigate the role of SES in modulating the relationship between blood lipids, metabolites, and metabolic disease outcomes.
4. To explore potential causal relationships using advanced statistical and machine learning techniques, such as Mendelian randomization and causal inference models.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/correlation-and-multiomics-studies-of-age-related-diseases-and-neurological-diseases

Correlation and multiomics studies of age-related diseases and neurological diseases

Last updated:
ID:
762894
Start date:
2 May 2025
Project status:
Current
Principal investigator:
Miss Xiaofang Yuan
Lead institution:
Third Affiliated Hospital of Soochow University, China

Research Question
Aging-related diseases (e.g., cardiovascular disorders, diabetes) and neurological disorders (e.g., Alzheimer’s disease, Parkinson’s disease) represent interconnected global health challenges. These conditions share molecular mechanisms including chronic inflammation, mitochondrial dysfunction, and protein aggregation, suggesting bidirectional links between systemic aging and neurodegeneration. Epidemiological evidence demonstrates that metabolic syndrome increases dementia risk, while neurodegenerative pathologies correlate with accelerated systemic aging. However, the molecular pathways linking these processes remain poorly understood, hindered by fragmented single-omics data and inadequate integration of modifiable risks with genetic susceptibility.
Objective
To address this gap, the present undertaking aims to employ an integrated approach, leveraging the wealth of genetics, metabolomics, proteomics, and comprehensive epidemiological data. Through multidimensional analyses, we aim to discover new risk factors, identify potential biomarkers, and understand causal relationships for various Neurodegenerative diseases.
Scientific rationale
Circulating proteins and blood metabolome serve as powerful biomarkers, integrating genetic, molecular, and environmental influences to elucidate disease etiology. We will combine multi-omics (i.e., proteomics and metabolomics) and epidemiological data to discover novel risk factors, biomarkers and provide definitive evidence for known associations of Neurodegenerative diseases risk reported by traditional observational studies. By integrating multi-omics (proteomics/metabolomics) with epidemiological data, we will identify novel risk biomarkers and decode causal mechanisms underlying neurodegenerative (e.g., Parkinson’s) and age-related diseases (e.g., stroke). This approach systematically addresses gene-environment interplay while validating observational associations through causal inference.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/correlation-between-genetic-heterozygosity-of-transcriptions-factors-and-large-artery-atherosclerosis-a-population-based-study

Correlation Between Genetic Heterozygosity of Transcriptions Factors and Large Artery Atherosclerosis: a Population-based Study

Last updated:
ID:
848357
Start date:
18 August 2025
Project status:
Current
Principal investigator:
Professor Qiaoshu Wang
Lead institution:
Shanghai General Hospital, China

Stroke is a leading cause of mortality worldwide. One of the most important etiologies of stroke is large artery atherosclerosis and genetics play an important role in the initiation and progression of atherosclerosis. Therefore, a deeper understanding of genetics that affecting atherosclerosis may lead to novel prevention and therapeutic strategies.

Several transcription factors have been identified to be associated with atherosclerosis, such as NF-kB, Krüppel-like factors, BACH1, etc. Those transcription factors modulate the activation of inflammatory mediators, thereby promoting the pathogenesis of atherosclerosis. Prior studies have characterized the role of transcription factors in rodent models, whereas the validation of the relationship between transcription factors and atherosclerosis in human is limited due to many reasons.

This project aims to systematically investigate the associations of genetic, hemodynamic profiles, as well as external environmental exposures with development of large artery atherosclerosis (carotid atherosclerosis, etc.) based on human biomedical data. The genetic heterozygosity of transcription factors established by animal models and their relationships with large artery atherosclerosis as well as ischemic strokes will be the main focus of this project. Leveraging population-level data from the UK Biobank, this project will contribute to novel insights into the identification of potential prevention and/or therapeutic targets for atherosclerosis.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/correlation-between-genetic-variants-diet-and-retinal-phenotype-in-age-related-macular-degeneration

Correlation between genetic variants, diet and retinal phenotype in age-related macular degeneration

Last updated:
ID:
92079
Start date:
16 March 2023
Project status:
Current
Principal investigator:
Dr Kanmin Xue
Lead institution:
University of Oxford, Great Britain

Age-related macular degeneration (AMD) is a leading cause of blindness in developed countries, predominantly affecting older people. This condition involves progressive damage to the central part of the retina (the tissue at the back of the eye that detects light) called the macula, which is responsible for our central vision and functions such as reading and seeing faces or fine detail. At the moment, treatment is only available for one type of AMD (“wet” AMD in which abnormal blood vessels grow under the retina). However, there is no treatment for patients with “dry” AMD and thus they are affected by progressing loss of vision. The causes of AMD remain incompletely understood, but appear to be a combination of genetic, dietary, and environmental factors which all affect the risk of an individual developing AMD and progressing to advanced stages of the disease.

The aim of this study is to improve our understanding of the causes of AMD, focusing on the effects of genetic risk factors and diet on AMD development. The study will take advance of comprehensive genetic, diet, blood, and retinal imaging data from the UK Biobank. Significant findings will be further validated in patients attending AMD eye clinics. Understanding how these factors interact with each other to increase a person’s risk of developing AMD will be crucial for devising preventative lifestyle measures and developing new treatments for this major cause of blindness.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/correlation-between-imaging-parameters-genetic-and-inflammatory-markers-for-diagnosis-and-treatment-prognosis-in-patients-with-osteoarthritis-and-cartilage-injury

Correlation Between Imaging parameters, Genetic and Inflammatory Markers for Diagnosis and Treatment Prognosis in Patients with Osteoarthritis and Cartilage Injury

Last updated:
ID:
351718
Start date:
29 October 2024
Project status:
Current
Principal investigator:
Miss Rafaella Rogatto de Faria
Lead institution:
University of Sao Paulo, Brazil

The main objective is to identify potential genetic markers, correlate with the inflammatory profile, imaging parameters and health status, and associate it with the clinical outcome for cartilage lesion and osteoarthritis. Several factors are implicated in the increased risk of osteoarthritis, such as age, overweight, trauma, surgical approach or congenital changes in the joint, gout, diabetes, and hormonal diseases.
Understanding the genetic variations present in people with cartilage damage and osteoarthritis, and how these patients respond to the proposed treatments, will bring benefits to the population, contribute to the advancement of national science and technology, and allow the choice of the most appropriate treatment, generating faster and more effective therapeutic effects. One of the tools that contribute to the development of personalized medicine are the Single Nucleotide Polymorphisms (SNPs).
However, there are no studies correlating SNPs with clinical outcomes in patients with chondral lesions who are candidates for surgical interventions directed at these lesions. In addition to genetic markers, blood biomarkers are studied as prognostic means for identifying individuals at high risk of progression of knee osteoarthritis.
There is a gap in the literature about this type of association, which could be used for precision medicine. It is believed that with an experimental analysis with SNPs, the dosage of inflammatory cytokines and the correlation with imaging parameters, health status and clinical outcomes, it will be possible to propose personalized and more assertive treatments to athletes and patients.
That said, the patients to be analyzed are those with cartilage damage and knee osteoarthritis, unresponsive to nonoperative therapy (strengthening, weight loss if applicable and guidelines for activities of daily living) with follow-up for a minimum period of 6 months and indication of surgical treatment (debridement, autologous chondrocyte implantation, osteochondral allograft surgeries and partial or total knee arthroplasty) according to the PROMS scales.
The initial part of the project consists of collecting biological samples and gathering the data available at public databases, such as UK Biobank, to start the validation and correlations, this part is estimated to last 14 months. In the remaining 22 months, a study will be carried out on the development of correlation between the parameters. Our group’s data will be validated using UK Biobank data as a population comparison. Information will also be stratified according to patients’ ancestry data and other available relevant data such as sex, weight and age to allow for adequate comparison of groups.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/correlation-of-periodontitis-with-morbidity-and-mortality-in-alzheimers-disease-and-its-molecular-mechanisms

Correlation of periodontitis with morbidity and mortality in Alzheimer’s disease and its molecular mechanisms

Last updated:
ID:
91625
Start date:
17 October 2022
Project status:
Current
Principal investigator:
Dr Hao Xu
Lead institution:
Sichuan University, China

To explore the relationship between periodontal disease (PD) and the morbidity and mortality of Alzheimer’s disease and its molecular mechanisms.
PD is one of the most common inflammatory diseases in humans and can cause destruction of soft and hard periodontal tissues, ultimately leading to tooth movement and loss. It is caused by a dysbiosis of the oral microbiota and is associated with a dysregulated immune inflammatory response. The response caused by bacterial build-up on the tooth surface is not confined to the mouth, but is also involved in the progression of systemic inflammation through the digestive tract or bloodstream.
Alzheimer’s disease (AD) is a chronic progressive neurodegenerative disease that occurs in middle-aged and elderly populations and is characterised by irreversible and progressive memory loss and cognitive impairment due to neuronal death. Current research suggests that there is a correlation between AD and PD. It has also been suggested that patients with AD often have poor periodontal health, leading to persistent infection of their periodontal tissues.The periodontal status of patients with AD is receiving increasing attention, particularly with regard to the relationship between periodontal status and the long-term health status of patients with AD.
Objective 1: To determine the correlation between periodontal disease (PD) and the morbidity and mortality of Alzheimer’s disease (AD)
To explore the correlation between PD and AD incidence using multifactorial logistic regression and the correlation between PD and mortality in AD patients using Cox’s regression.
Objective 2: Identification of new genetic and non-genetic determinants
For this section, genetic and phenotypic correlations between PD and AD will be estimated to identify shared pathways associated with both diseases. We will also target specific genetic and other markers for candidate association analyses based on prior biological and clinical knowledge.
Objective 3: Critical assessment and causal inference
Mendelian randomisation analyses were performed and causal inferences were made for evidence with high confidence levels. Chain disequilibrium score (LDSC) regression and Mendelian randomisation (Mr) analyses allow further analysis of the relationship between PD and AD.
Combining the large data available from the UK Biobank with our analysis of epidemiological and molecular mechanisms will add to the current understanding of the pathogenesis and progression of PD and Alzheimer’s disease. Our findings may also provide evidence for personalised disease prevention and management. This project is likely to extend beyond three years as the newly released data will require additional validation of our findings.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/correlation-study-of-double-gene-mutations-with-the-onset-of-premature-ovarian-insufficiency

Correlation study of double gene mutations with the onset of Premature Ovarian Insufficiency

Last updated:
ID:
177789
Start date:
13 May 2024
Project status:
Current
Principal investigator:
Ms Yuxi Ding
Lead institution:
Tsinghua University, China

Infertility affects an estimated 48 million couples and 186 million individuals worldwide. Premature ovarian insufficiency (POI), alongside generalized late marriage and childbirth, is one among many factors contributing to infertility. The causes of POI are heterogeneous, and genetic abnormalities, including chromosome number abnormalities and single-gene mutations, account for up to 30% of all POI cases. However, recent studies have suggested that the predictive power of individual gene mutations for the onset of premature ovarian insufficiency is weak, and the collaborative pathogenecity of double or multiple mutations is instead more likely to cause the phenotype in POI individuals. Nevertheless, current experiences about double mutations in POI individuals or families remain relatively limited.
We identified a family with a Premature ovarian insufficiency (POI) history. The family has several POI individuals. However, by whole exon sequencing(WES), we didn’t identify a single gene mutation that was directly associated with the disease phenotype. Instead, we find that the affected individuals share two mutations in which one is more scarce and the other is more common(frequency > 0.1%) among the general population. We suppose the two mutations might act synergistically to cause the POI phenotype. To verify this hypothesis, we want to use the UK biobank to examine whether this mutation pair is significantly associated with early menopause.
The project will last for 36 months.The investigation aims to elucidate whether the co-occurrence of these mutations is significantly associated with early menopause. This approach recognizes the complexity of genetic contributions to POI, moving beyond the traditional focus on single-gene mutations. By investigating the correlation of paired gene mutations with female reproductive longevity, we will contribute to current understanding of the genetics of POI, and help to reveal new therapeutic targets for infertility.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cortex-map-a-web-service-for-mri-based-localization-of-brain-abnormalities-in-epilepsy

Cortex.Map: a web service for MRI-based localization of brain abnormalities in epilepsy

Last updated:
ID:
36080
Start date:
21 February 2019
Project status:
Closed
Principal investigator:
Dr Heath Pardoe
Lead institution:
NYU Grossman School of Medicine, United States of America

The aim of this study is to develop a web-based tool for mapping brain abnormalities in individuals using neuroanatomical MRI. This tool will be useful for localizing cortical changes in epilepsy patients, and will be useful for presurgical planning in individuals with medically intractable epilepsy. Imaging data collected as part of the UK Biobank study will be combined with other large neuroimaging datasets to generate a large population-level database of normative quantitative imaging metrics that can then be used to identify subtle brain changes in individual epilepsy patients. The outcomes of our study will improve epilepsy diagnosis and treatment. Techniques developed as part of the proposed study will also be useful for similar web-based tools in other neurological disorders, and will therefore contribute to the stated purpose of improving diagnosis and treatment of illness, with a specific focus on neurological disorders. We will build a web-based tool that allows users to upload brain MRI scans of epilepsy patients, and obtain maps that show where the brain structure in these patients is abnormal. This often indicates where seizures are coming from in the brains of people with epilepsy. In patients with severe epilepsy, brain surgery is often the only effective treatment option. The Cortex.Map tool will assist surgeons to plan which brain regions to remove to stop these patients having seizures. UK Biobank participants with brain MRI scans will be included in this study. Therefore approximately 10,000 participants will be included in our study.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cortical-genotype-phenotype-influences-on-inter-individual-variability-in-neurodegeneration

Cortical genotype-phenotype influences on inter-individual variability in neurodegeneration

Last updated:
ID:
30730
Start date:
15 March 2018
Project status:
Closed
Principal investigator:
Christian Lambert
Lead institution:
City St George's, University of London, Great Britain

The causes of variability between individuals in brain function and disease are complex. Some of the differences may be due to differences in brain anatomy, for example the structure of someone?s brain may predispose them to more severe symptoms through disease related damage. Parkinson?s disease is a common condition well suited to study this question. We aim to: A) Understand whether genes associated with higher risk of PD modify the structure of the brain in healthy individuals. B) Whether structural brain changes, which we have previously characterised in PD and pre-clinical dementia, have any genetic associations. These questions are designed to provide new insights into the interaction between genes, environment and brain structure. The objective driving these questions is to develop ways of improving precision medicine. By understanding how these factors alter the structure of the brain in the normal population, this project will contribute to ongoing work into detecting pre-clinical disease and predicting how these conditions will progress in individuals. This work will also identify and quantify individuals ?at risk? of Parkinson?s disease, and we have local institutional support to follow these up and later assess the longitudinal health care outcomes. Using structural MRI, genetic, demographic and cognitive data acquired for 100,000 healthy individuals:

Question 1:
All individuals who have common genetic variants associated with an increased risk of either developing PD, or progressing more quickly once it has manifested, will be identified. Using these subgroups, differences in brain structure and physical characteristics will be identified. If there are significant differences in brain structure, these measures will be used to try and identify related genes.

Question 2:
The similarity between healthy and diseased brains will be quantified, and used to measure differences between groups, and to search for genetic associations. This work will focus on individuals with no pre-existing neurological disease. We have requested the maximum available MRI dataset to provide sufficient frequencies of the outcomes of interest (see below)

Using the full dataset will allow the MRI derived metrics to be used in genome wide association analyses using pathological patterns of disease. This latter objective will identify other genetic variants that modulate anatomical structures involved in disease and may therefore contribute to the observed pathological variability.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/create-multi-modal-brain-image-priors-for-use-in-bayesian-models-to-be-applied-in-stroke-lesion-segmentation-and-ad-diagnosis-studies

Create multi-modal brain image priors for use in Bayesian models to be applied in stroke lesion segmentation and AD diagnosis studies

Last updated:
ID:
93346
Start date:
28 November 2022
Project status:
Current
Principal investigator:
Professor Yao Li
Lead institution:
Shanghai Jiao Tong University, China

Stroke and neurodegenerative disorders such as Alzheimer’s disease are among the leading causes of death worldwide and have imposed a heavy economic burden on society. The development of imaging-based biomarkers for the early and accurate diagnosis of them is of critical clinical need. In this research, we seek to: (1) characterize the normal brain features using the UK Biobank imaging data; (2) develop a lesion detection algorithm that separates the lesion tissue from normal brain tissue using Biobank image data; (3) develop an algorithm for the early detection of Alzheimer’s disease using Biobank image data; (4) Investigate the influence of genetic and environmental risk factors to the variations in brain features.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/creating-a-pancreatic-cancer-control-group-for-a-pancreatic-cancer-project-cru5002

Creating a Pancreatic Cancer Control Group for a Pancreatic Cancer Project (CRU5002)

Last updated:
ID:
100681
Start date:
9 January 2025
Project status:
Current
Principal investigator:
Professor Ulrich Sax
Lead institution:
University Medical Center Göttingen, Germany

[Aims] Pancreatic cancer is a severe disease that can lead to death within a few month in many patients. Pancreatic ductal adenocarcinoma (PDAC) is a difficult cancer to treat. Recent research has shown that PDAC has different subtypes, each with different characteristics that affect the way the cancer grows and responds to treatment. These differences are due to changes in the cancer’s genetic makeup. Understanding these changes can lead to better treatment strategies based on a patient’s subtype, which is known as precision medicine.
[Rationale] The Clinical Research Unit (CRU 5002) at the University Medical Center Göttingen aims to understand how genetic changes drive subtype-specificity in PDAC. The unit will use advanced tumor models and sequencing technologies to study seven different projects related to PDAC subtypes.
Unfortunately, there are not many datasets available documenting Pancreatic cancer in huge cohorts.The UK Biobank data set could help to widen our local data sets. Our team will also compare their findings to data from a large group of PDAC patients in the UK Biobank.
This comparison will help them find therapies specific to each subtype and improve treatment outcomes for future patients.
[Expected duration of project] the overall project will take 36 month, as it includes studies on mouse models and cell lines as well additional to the here described data projects.
[public health impact] Pancreatic cancer currently accounts for about 5% of all cancer deaths. The risk to die of pancreatic cancer will rise over the next years, So understanding the risk factors leading to pancreatic cancer and finding the correct treatment for each kind of pancreatic cancer is very important for society and for public health.
Overall, this research is important for improving treatment strategies for PDAC patients and advancing precision medicine.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/creating-and-evaluating-polygenic-scores-for-risk-and-response-for-common-complex-chronic-diseases-in-multiple-populations

Creating and evaluating polygenic scores for risk and response for common complex chronic diseases in multiple populations

Last updated:
ID:
63258
Start date:
9 November 2020
Project status:
Current
Principal investigator:
Professor David Ethan Lanfear
Lead institution:
Henry Ford Hospital, United States of America

Chronic diseases like heart diseases, diabetes, and obesity are leading causes of death in the U.S. Genetic risk scores can identify risks for disease independent of traditional clinical risk factors. Another important issue is that polygenic risk scores have largely been developed in European population groups, and therefore, do not perform well in non-white population group.
The main goal of this project is to develop genetic risk scores that can be used to help predict a person’s risk of a condition, and response to drug treatment. Ancestry-specific risk scores will be created and evaluated in non-white population groups, utilizing effect estimates generated in European and African-ancestry population groups. This work will improve the individualization of risk scores and, therefore, the application of precision treatments. We expect to complete the proposed work within 36 months. At the end of the proposed work, we expect to have validated risk prediction model on a few complex diseases and their related drug response phenotypes for multiple populations. The potential public health impact is enormous because the diseases of interest are very common and quite morbid, and we currently have inadequate tools to predict who will develop the disease or whether an individual will respond to the usual treatments. This is particularly true of non-white ancestral groups for whom the genetic factors influencing disease and drug response are less explored. For example current guidelines suggest treating all heart failure patients with Beta blockers but our preliminary data suggests that most of this benefit occurs in only one-third of patients with the other two-thirds deriving little benefit. If polygenic scores can better direct our preventative and treatment efforts to the subgroups of patients at highest risk or with enhanced benefit, this would revolutionize medical care and make it much more efficient.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/creating-and-validating-integrated-risk-scores-that-incorporate-polygenic-risk-scores-rare-variant-burden-and-family-history

Creating and validating Integrated Risk Scores that incorporate polygenic risk scores, rare variant burden, and family history

Last updated:
ID:
680373
Start date:
10 March 2025
Project status:
Current
Principal investigator:
Dr Elan Bechor
Lead institution:
Minerva Genomics Inc., United States of America

Our objective in this study is to build and validate an “Integrated Risk Score” for diseases and traits, defined to be a single prediction function (such as a logistic regression) containing a polygenic risk score, family history information, and the predicted effects from rare variants. We want to validate these scores across ancestries and within families to test the robustness of the scores.

The research will involve analyzing multiple segments of the UK Biobank’s data, including the rare variants extracted from the whole genome sequencing data, family history information from the survey data, and genotypes from the imputed genotypes. The project will conclude by creating prediction models for diseases and traits in the form of an Integrated Risk Score, which combines all three sources.

Research questions:

1. Can the trans-ethnic portability of polygenic risk scores be improved by integrating data from diverse ancestries and using techniques such as X-Wing (Miao 2023), SBayesRC (Zheng 2024)?
2. Are these resulting polygenic risk scores attenuated within families by e.g. assortative mating (Young 2023)?
3. Can rare variants from exome wide association studies yield incremental preedictive value? Are these trans-ethnically portable?
4. Can we integrate the rare variants, polygenic scores and family history information in a way grounded by mathematical theory?

Scientific Rationale:

To our knowledge, this Integrated Risk Score would be the first of its kind and would contribute to the literature. We are only aware of studies that incorporate two of the three. For example, rare and common variants have been analyzed together in the case of type 1 diabetes (Dornbos 2023). Family history and polygenic scores have been integrated, e.g. (Mars 2022).


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/creation-of-a-framework-for-identification-of-patterns-of-brain-health-and-disease

Creation of a framework for identification of patterns of brain health and disease

Last updated:
ID:
41865
Start date:
12 March 2019
Project status:
Closed
Principal investigator:
Dr Gabriel Gonzalez-Escamilla
Lead institution:
UMC of the Johannes Gutenberg University, Germany

The most important challenges in clinical and biomedical research include the need to develop and apply tools for the effective integration, analysis and interpretation of complex data with the aim to identify testable hypothesis, and build accurate models of disease development and progression.
Recent advances in magnetic resonance imaging techniques allow the acquisition of increased amounts of data. With more and more data available, machine learning techniques, which are closely related to statistics, are becoming increasingly popular. Machine learning allows predictions based on sets of individual variables. The use of Machine Learning, more superficially deep learning, in Healthcare problems can be of great importance, mostly because it offers the opportunity of developing algorithms that identify complex patterns within large amounts of data, otherwise unachievable with standard methods.
Health maintenance is in fact a multifactorial phenomenon, determined by interactions of its factors, including genetic inheritance, internal physiological processes, personal behaviors, and the general external environment. Furthermore, neurodegenerative diseases are neurological illnesses that manifest as movement disorder, cognitive impairment, and/or psychiatric disturbance, attributed to neuronal cell death. Alzheimer’s disease (AD), Parkinson’s disease (PD), among others are the best known neurodegenerative diseases, but also demyelinating diseases, such as multiple sclerosis belong to this category. These illnesses occur worldwide representing a great challenge and effort for healthcare systems. We propose that by feeding a deep learning algorithm with several amount of data from the UK biobank the deep learning algorithm can learn how to recognize patterns associated and differentiated with certain phenotypes, such as disease conditions (e.g., multiple sclerosis). The definition of a framework for correct and opportune disease detection and surveillance would provide way for epidemiologic studies that facilitate health experts to deploy preventive measures and help healthcare administrators to make optimal decisions.
Further, such a context provides a novel way to capture individual differences within the general population as well as disease groups that relate to additional facets of various diseases or even predict future outcomes.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cross-cultural-variations-in-female-reproductive-health-and-non-communicable-diseases-analysis-of-500000-participants-from-the-ukbiobank-and-the-interlace-consortium

Cross-cultural variations in female reproductive health and non-communicable diseases: analysis of 500,000 participants from the UKBiobank and the InterLACE consortium

Last updated:
ID:
26629
Start date:
1 June 2017
Project status:
Current
Principal investigator:
Professor Gita Mishra
Lead institution:
University of Queensland, Australia

The aim of this study is to incorporate Biobank data as a major addition to the International Collaboration for a Life course Approach to reproductive health Chronic disease Events (InterLACE), a leading international research collaboration. Established in 2012, the focus of InterLACE is on understanding women?s reproductive characteristics and links with the risks of key non-communicable diseases (NCDs), such as cardiovascular disease (CVD), type 2 diabetes, and depression. InterLACE is currently using data from over 230,000 women in 20 studies from nine countries, including Sweden, the USA, Australia, and Japan. Due to the sample size and quality of data collected, the addition of Biobank data with greatly strengthen and expand the research capability of InterLACE. The proposed project is health-related and is in the public interest. Findings will enable the use of reproductive factors as part of an integrated approach to the development of timely and targeted preventive health strategies, such as medical surveillance, to reduce the risk of NCDs. This project will utilise individual-level data from both the UK Biobank and the InterLACE dataset ? see above.

Generalised Estimating Equations, multinomial logistic regression, will be used to determine the associations between reproductive factors and and the incidence of NCDs.
We would require information only from female participants


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cross-diagnostic-and-cross-platform-multimodal-analysis-of-uk-biobank-imaging-data

Cross-diagnostic and cross-platform multimodal analysis of UK Biobank imaging data

Last updated:
ID:
47267
Start date:
25 March 2019
Project status:
Current
Principal investigator:
Dr Janine Diane Bijsterbosch
Lead institution:
Washington University in St. Louis, United States of America

MRI scanners have the potential to help doctors decide whether we are at risk of certain diseases. To reach this potential, we first need to study a very large number of people to understand which MRI-based measures are most informative. We also need to make sure that we study people from different countries and across different ages to make sure that the MRI-based predictions can be used for everyone. This project brings together several big research studies so that we can look at the same MRI-based measures across continents, ages, and groups to find the best MRI-based predictors of disease.

Our aim is to compare MRI-based markers of disease between the UK Biobank and other big data initiatives such as the Human Connectome Project, Connectomes Related to Human Disease projects, and the “All of us” study. We will analyse the MRI data to extract potential markers (for example based on connectivity patterns in the brain), and to develop new markers. We want to understand what these MRI markers can potentially tell us about health outcomes, and how they are related to other aspects of our lifestyle and our body’s physiology. We will use advanced analysis methods and develop new approaches to study the complex relationships between MRI-based measures and other factors such as genetics, lifestyle, and physiology, in a range of psychiatric and neurodegenerative diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cross-disease-analysis-of-shared-and-unique-influence-factors-a-comprehensive-study

Cross-Disease Analysis of Shared and Unique Influence Factors: A Comprehensive Study

Last updated:
ID:
142341
Start date:
31 July 2025
Project status:
Current
Principal investigator:
Professor Sanbing Shen
Lead institution:
University of Galway, Ireland

Diseases often have underlying causes or factors that contribute to their onset. We aim to uncover these triggers, focusing on those that might be common to multiple diseases, as well as those specific to individual ailments. Think of it like identifying shared and unique ingredients in various recipes.
Usually, researchers tackle diseases one at a time. However, if several diseases share common triggers, understanding these can revolutionize prevention and treatment. For instance, if one lifestyle choice increases the risk for multiple diseases, addressing it can combat several health issues at once.
We’re using data from the UK Biobank, which contains information from hundreds of thousands of people, covering aspects like genes, diagnosed diseases, lifestyles, and environmental factors.
Our approach is threefold: Identify factors linked to individual diseases; Group diseases by their shared and unique factors; Ensure our results aren’t skewed by unrelated factors, such as age or gender.
This comprehensive study will span several years. Given the vast data and variety of diseases we’re examining, this timeframe ensures accurate and thorough findings.
The implications for public health are significant. Recognizing common disease triggers can lead to:
We’re delving deep into data to pinpoint shared and distinct triggers for various diseases. This research has the potential to reshape health strategies, offering more targeted prevention, personalized treatments, and efficient use of health resources.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cross-disease-biomarker-analysis-of-alzheimers-disease-for-identifying-shared-pathological-pathways-and-optimal-timing-for-diagnostic-assessments

Cross-Disease Biomarker Analysis of Alzheimer’s Disease for Identifying Shared Pathological Pathways and Optimal Timing for Diagnostic Assessments

Last updated:
ID:
469501
Start date:
4 February 2025
Project status:
Current
Principal investigator:
Dr Adam Stephens
Lead institution:
Cytocell Ltd, Great Britain

The proposed research aims to investigate the correlation between Alzheimer’s disease-specific biomarkers and those associated with other major diseases, including cancer and neurodegenerative disorders, to identify shared pathological pathways and optimal diagnostic timing. Using data from a cohort of 10,000-15,000 participants from the UK Biobank, the study will employ a comprehensive analysis of biomarker data, including protein levels, reported genetic markers, and patient records.
The research will utilise statistical models, such as multivariate regression and principal component analysis, to identify significant relationships between Alzheimer’s biomarkers and those of other diseases. Additionally, machine learning models, including clustering algorithms and random forests, will be applied to uncover common biomarker profiles across diseases and determine the most relevant clinical timepoints for biomarker assessment. Time-series and survival analysis will be conducted to correlate biomarker expression with disease onset and progression.
The goal is to refine diagnostic strategies by identifying shared biomarker signatures and optimal assessment timings, potentially leading to earlier detection and more personalised treatment strategies. The findings from this research could significantly impact public health by improving diagnostic practices and outcomes for patients with multi-morbidity.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cross-influence-of-shared-risk-factors-in-cardiovascular-disease-and-cancer

Cross-Influence of Shared Risk Factors in Cardiovascular Disease and Cancer

Last updated:
ID:
633186
Start date:
9 May 2025
Project status:
Current
Principal investigator:
Dr Qinghua Yuan
Lead institution:
The Seventh Affiliated Hospital of Sun Yat sen University, China

Cardiovascular diseases represent one of the leading causes of mortality globally. They account for a significant proportion of morbidity and mortality in many countries. Due to the adoption of Westernized lifestyles witnessed a rapid increase in cardiovascular disease incidence. Cardiovascular diseases are closely associated with hypertension, hyperlipidemia, hyperglycemia (“three highs”), smoking, obesity, physical inactivity, age,gender and psychological stress. These risk factors often coexist and synergistically elevate the risk of cardiovascular diseases.
Global cancer incidence has been increasing annually. A tumor is an abnormal growth resulting from uncontrolled cellular proliferation under the influence of various carcinogenic factors. This trend is linked to population aging, environmental pollution, and lifestyle changes (such as smoking, alcohol consumption, high-calorie diets, and sedentary behavior). At the genetic level, local tissue cells lose normal growth regulation, leading to clonal dysplasia. Tumors can be classified into benign and malignant types. Malignant tumors, including carcinomas (e.g., lung, gastric, breast cancer) and sarcomas, exhibit invasive and metastatic properties, causing severe damage to other tissues and organs.
Tumor types are diverse, with varying incidence and mortality rates across different organs. Most tumor incidences increase with age, with a higher risk observed in elderly populations. Some tumors exhibit gender-specific patterns; for example, men have a higher incidence of lung and stomach cancers, while women are more prone to breast and cervical cancers.
Investigating the Molecular Mechanisms of Shared Risk Factors in Cardiovascular Diseases and Cancers, Common risk factors activate intracellular oxidative stress responses, generating excess reactive oxygen species (ROS).


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cross-modal-medical-analysis-and-reasoning

Cross-modal medical analysis and reasoning

Last updated:
ID:
70340
Start date:
26 August 2022
Project status:
Current
Principal investigator:
Professor Wensheng Zhang
Lead institution:
Institute of Automation (Chinese Academy Of Sciences), China

The cross-modal medical knowledge graph (a collection like a network that contained relationships between different types of medical data, e.g., images, text, etc.) is becoming critical for both academic research and clinical applications. Generating a representation that could reflect different modalities (organized by knowledge graph) remains challenging and underdeveloped. We will a) rely on the existing medical expert knowledge of the clinical disease to construct the basic structure for the knowledge graph, b) use statistic models to induce the features and relations from different data modalities in the UKBB and incorporate with the basic structure, c) use optimization algorithms to refine the structure of knowledge graph, d) use artificial intelligence methods to compute the similarity between two patients, and e) find the similar patient according to the clinical requirement. The UKBB data is essential to our knowledge graph construction because of its rich information from multiple modalities. Moreover, we will adjust the structure of the knowledge graph according to the updates in the field of medical science. Besides, we will also study the approaches to explain the process of computer reasoning. The project duration is 3 years. It can support the clinical decision, narrow the gap of medical resources between regions, improve the overall medical service level. In addition, UKBB data will also be used as a benchmark dataset for evaluating the related methods of the cross-modal medical knowledge graph.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cross-modality-cardiac-scar-imaging

Cross-Modality Cardiac Scar Imaging

Last updated:
ID:
49156
Start date:
7 May 2019
Project status:
Closed
Principal investigator:
Steven Niederer
Lead institution:
King's College London, Great Britain

Imaging cardiac scar tissue is important to assist in the diagnosis and treatment of many heart conditions. The current standard method is an MRI scan with gadolinium contrast agent. Many patients, such as those with kidney disease, are unable to get these scans due to the pressure it puts on the renal system. Others with cardiac implants such as pacemakers cannot receive it due to image artefacts from the metal in the implant interfering with the scanner.

This study aims to use data from patients who have had cardiac MRI scans to develop an algorithm which can predict scar in the heart wall without contrast agent. Scar tissue presence causes differences in shape and density of heart tissue which are difficult to manually identify but can be automatically identified using machine learning methods. Detecting these signs of scar without contrast agent would therefore be possible. We aim to make this method viable with other scanning methods such as CT or echocardiography since they also would be able to detect the signs of scar from the shape and density of the heart wall tissue. Since these scans are routinely carried out in cardiac clinics the clinicians would receive additional information from scans the patient is already receiving using this method. Such a method could be used to inform whether the patient would require an MRI or further clinical follow up.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cross-omics-studies-on-endocrine-and-metabolic-system-diseases-and-multisystem-complications-a-comprehensive-analysis-from-mechanism-to-clinical-application

Cross-Omics Studies on Endocrine and Metabolic System Diseases and Multisystem Complications: A Comprehensive Analysis from Mechanism to Clinical Application

Last updated:
ID:
511414
Start date:
14 April 2025
Project status:
Current
Principal investigator:
Professor Guixia Wang
Lead institution:
First Hospital of Jilin University, China

1. Background
Endocrine and metabolic system diseases, such as diabetes, obesity, hyperlipidemia, hyperuricemia, bone metabolism-related diseases, pituitary diseases, adrenal diseases, thyroid diseases, and parathyroid diseases, not only pose direct health risks to patients, but also frequently lead to cardiovascular diseases , kidney diseases, neurological disorders, and digestive system diseases, causing multisystem complications. These complications significantly increase the complexity of patient management,not only severely impacting patients’ quality of life,but also heightening their risk of mortality.Moreover, they present a substantial challenge to global public health.
2. Objectives
The primary objective of this study is to elucidate the clinical characteristics and molecular mechanisms of endocrine metabolic diseases and their associated multisystem complications.
3. Study Design and Methods
This study is a prospective cohort study utilizing a cohort population from an established database. We screened patients with endocrine and metabolic diseases that met the study criteria, and analyze the data using methods such as Cox regression analysis.
4. Expected Outcomes
We expect this study to clarify the clinical associations and pathogenesis of endocrine metabolic diseases and their complications, identify new biomarkers and potential therapeutic targets, and provide more precise diagnostic and therapeutic strategies for clinical practice, thereby improving patient prognosis and quality of life.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cross-race-examination-of-obesity-risk-prediction-through-genome-wide-association-study-and-polygenic-risk-score-development

Cross-Race Examination of Obesity Risk Prediction through Genome-Wide Association Study and Polygenic Risk Score Development

Last updated:
ID:
301868
Start date:
24 October 2024
Project status:
Current
Principal investigator:
Miss Kezia Irene
Lead institution:
PT Kalbe Farma Tbk, Indonesia

Obesity is a global epidemic, with its prevalence increasing rapidly, especially in regions like Indonesia. While factors like diet and exercise play a role, recent research suggests genetics also influence obesity risk. However, most genetic studies on obesity have focused on European populations, raising questions about their relevance to diverse groups, including Asians. Our project aims to bridge this gap by studying the genetics of obesity across different racial and ethnic groups, particularly in Southeast Asia.

We aim to uncover the genetic factors contributing to obesity susceptibility across diverse populations, including Indonesians. By leveraging data from the UK Biobank, which includes diverse populations, and combining it with our in-house genetic data from Indonesians, we aim to identify genetic markers specific to Asian populations, shedding light on why certain groups may be more susceptible to obesity than others. By doing so, we hope to identify genetic factors specific to Asian populations, shedding light on why certain groups may be more susceptible to obesity than others. This information could lead to tailored interventions and healthcare strategies, particularly beneficial for countries like Indonesia where obesity rates are rising.

Our study will involve advanced genetic analyses to uncover key genetic markers associated with obesity risk. We’ll also explore how these genetic factors interact with lifestyle and environmental factors, providing a comprehensive understanding of obesity’s genetic underpinnings. Importantly, we’ll ensure our research is ethical and inclusive, considering the needs of all populations, including minority groups often underrepresented in genetic studies.

The study duration is expected to be 36 months. Furthermore, our study aims to have a lasting impact that extends beyond the immediate project duration. Our study’s outcomes will inform Indonesian governmental policies aimed at preventing obesity from an early age, particularly if we find higher obesity risks within the Indonesian population. These policies might include additional mandatory teaching materials in schools, raising awareness about behaviors that can trigger obesity. Additionally, by pioneering comprehensive genetic research in Indonesia, our work can catalyze further interest and investment in genomic studies, particularly those utilizing Genome-Wide Association Studies (GWAS). Establishing research sustainability in genomics within Indonesia is crucial, not only for addressing obesity but also for advancing healthcare strategies tailored to the unique genetic makeup of Indonesian populations. This, in turn, could lead to more effective interventions and policies aimed at curbing the obesity epidemic and improving public health outcomes in Indonesia and beyond.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cross-sectional-study-exploring-the-effect-of-the-atg16l1-rs2241880-snp-t300a-on-respiratory-infection-incidence-respiratory-and-cardiovascular-health-of-the-uk-biobank-population

Cross sectional study exploring the effect of the Atg16L1 rs2241880 SNP (T300A) on respiratory infection incidence, respiratory and cardiovascular health of the UK Biobank population.

Last updated:
ID:
105295
Start date:
31 August 2023
Project status:
Current
Principal investigator:
Miss Maria Joao Ramos
Lead institution:
Quadram Institute Bioscience, Great Britain

We aim to find out if people carrying small mutations, known as single nucleotide polymorphisms or SNPs, have an increased risk of responding badly to respiratory infections and if this has an impact on overall respiratory health. The recent SARS-CoV-2 pandemic has highlighted the high social and economic cost of respiratory viruses, and thus the urgent need to be able to identify people at higher risk so that they can receive priority for treatment

The gene I’m interested is called Atg16L1 and has an important role in fighting off microbial infections. In mice, ATG16L1 has been shown to protect against acute influenza virus infection, by preventing virus from escaping into the lungs where it causes inflammation. It is known that mutations in Atg16L1 increase susceptibility of humans to inflammation of the gut during Crohn’s disease We think mutations in Atg16L1 could also affect the ability of humans to control inflammation during respiratory infection. Currently most studies on individuals carrying the Atg16L1 SNP are focused on Crohn’s disease and gut microbiome, and there is limited to no data on human studies looking at viral and bacterial respiratory infection incidence, or how this SNP affects the overall respiratory and cardiovascular health in these individuals.

With this project, I want to explore if people with a SNP in Atg16L1 have increased risk of worse outcomes following viral and bacterial pneumonia, and if this leads to worse overall respiratory and cardiovascular health. If this proves to be the case, people that have these SNPs can be identified and offered early access to vaccinations and be given priority and earlier treatment if required. For the NHS, early or preventive treatment will reduce how long a person will have to stay in hospital and can save the NHS both time and money.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cross-sectional-study-of-genome-wide-associations-with-fertility

Cross-sectional study of genome-wide associations with fertility

Last updated:
ID:
156667
Start date:
31 January 2024
Project status:
Current
Principal investigator:
Mr Daniel Ringwalt
Lead institution:
Johns Hopkins University, United States of America

Human fertility is tracked at the population level, often by survey participants reporting whether they have biological children, and the number of such children. Population averages of fertility are often reported at the national level, and as individuals’ social influences and plans for a family are not collected, we model the entire range of statuses (as a biological mother or father of one or more children) among a nation-scale population. Our project is a genome-wide association study with reproduction in the adult UK population. The project differs from the existing genome-wide association study, which associated each genotype with number of biological children in male and female cohorts, without regard to age. In the set of UK Biobank participants, who cover a cross-section of time, reproductive outcomes are modeled at a continuous rate, in cohorts grouped by both sex and age. For both sexes and at each age interval (a cohort), we fit average number of new children, as well as the entire range of outcomes at this particular age. The variability of outcomes in each cohort is driven by individual choice, and the average and spread of outcomes differ in each cohort. Significant genotype associations with a perturbed reproductive rate (in any cohort) might be driven by changes in fertility, driving the number of children lower for one genotype. The association tests will generate new hypotheses for genes that could interfere with, or protect, fertility. In public health, gene expression may be impacted by environmental factors, and may also be applied as a drug target. The former considerations can be part of evidence-based reproductive health care, while we would also like to make fertility extension possible by suggesting potential targets in pharmacology.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cross-sectional-study-to-investigate-ethnic-differences-in-cardiovascular-risk-and-mental-health

Cross-sectional study to investigate ethnic differences in cardiovascular risk and mental health

Last updated:
ID:
774
Start date:
5 November 2012
Project status:
Closed
Principal investigator:
Professor Jill Pell
Lead institution:
University of Glasgow, Great Britain

One advantage of UK Biobank is the recruitment of participants from ethnic minority groups in sufficient numbers to enable meaningful comparisons of different ethnic groups. Ethnic groups are known to differ in their risk of a number of conditions including cardiovascular disease and mental health. For example Pakistani people have a high risk of heart disease and chinese people a high risk of high blood pressure and stroke. Understanding these differences and the reasons for them is of assistance in ensuring the appropriateness and effectiveness of screening, investigation and treatment interventions.

The aim of this study is to compare the different ethnic minority groups in terms of the amount and type of disease, the distribution by age, sex and socioeconomic deprivation and the lifestyle and environmental factors that are associated with the presence of disease.

In this study we will access only questionnaire and measurement data and compare ethnic sub-groups in terms of these data. At a later date, once available, we will be able to compare these sub-groups in terms of their biochemistry assays and follow-up events. Therefore, this initial study will focus on comparisons of risk and only later will we be able to make comparisons of actual disease occurrence.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cross-sectional-vs-longitudinal-aging

Cross-sectional vs. Longitudinal Aging

Last updated:
ID:
117074
Start date:
4 October 2023
Project status:
Current
Principal investigator:
Ms Zoya Mooraj
Lead institution:
Max Planck Institute for Human Development, Germany

Aging is an important field of study within cognitive neuroscience, and this research into understanding how aspects of brain structure and function change with age is crucial to better understand the process of healthy aging, and to track how individuals fare in this process. However, much aging research is done cross-sectionally, taking measurements from only one timepoint into consideration, or by investigating group differences between a group of younger and a group of older adults.

As many others have done in the past, we argue that studying aging in this single snapshot manner does not accurately depict aging-related neural changes, especially in comparison to longitudinal studies which are able to accurately track within-person effects, thereby better capturing the nature of the change of brain and cognition as a person ages. With this work, we aim to take this a crucial step further by showing that a) longitudinal studies are more effective than cross-sectional studies at capturing “true” age-related changes, and b) how and why this relationship is necessarily constrained by the way in which measures of change are calculated. The project will take likely take 36 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cumulative-evidence-for-causal-relationships-between-extrinsic-exposures-and-major-noncommunicable-diseases

Cumulative evidence for causal relationships between extrinsic exposures and major noncommunicable diseases

Last updated:
ID:
48326
Start date:
13 March 2019
Project status:
Current
Principal investigator:
Professor Ben Zhang
Lead institution:
Sichuan University, China

Noncommunicable diseases account for more than 70% of the total human deaths worldwide. Cardiovascular diseases, cancers, chronic respiratory diseases, and diabetes are the four major types of noncommunicable diseases. Extrinsic exposures including but not limited to environmental, behavioral, occupational, lifestyle, metabolic factors have a major role in the development of most types of noncommunicable diseases. Over the past century, epidemiological studies have identified many risk factors for noncommunicable diseases. Recently, several large randomized controlled trials designed to evaluate the efficacy of new therapies targeted at well-established risk factors for noncommunicable diseases, however, have reported lower benefits than expected. Subsequent observational study of the same trial data has not clarified these unexpected findings. Thus, it is important and necessary to determine whether these well-established risk factors are causally associated with noncommunicable diseases before randomized controlled trials are conducted. In this application, we aim to systematically investigate associations between extrinsic exposures and risk of major noncommunicable diseases and provide cumulative evidence for causal relationships between well-established risk factors and major noncommunicable diseases. To address these issues, we will carry out a meta-analysis of prospective observational studies, a phenome-wide mendelian randomization analysis, and a meta-analysis of randomized clinical trials using data from UK Biobank, public data sources, and published studies. We will start analyses as soon as data are available and plan to finish this project and send manuscripts to authors for review within 36 months after we receive the data. We hope that our study will provide cumulative evidence for causal relationships between extrinsic exposures and risk of major noncommunicable diseases, and may help identify novel therapeutic targets for improving prevention and treatment of these complex diseases. Our study is consistent with the goal of UK Biobank that devotes to improving the prevention, diagnosis and treatment of a wide range of serious and life-threatening illnesses like major noncommunicable diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/cytomegalovirus-infection-and-risk-of-age-related-macular-degeneration-in-the-uk-biobank-cohort

Cytomegalovirus infection and risk of age-related macular degeneration in the UK Biobank cohort.

Last updated:
ID:
532689
Start date:
7 March 2025
Project status:
Current
Principal investigator:
Dr Morgane Linard
Lead institution:
University of Bordeaux, France

Age-related macular degeneration (AMD) is a neurodegenerative disease affecting the retina, and the leading cause of vision loss in the elderly in developed countries. Although several risk factors have been identified, the factors triggering the disease are still not fully understood.
Among existing hypotheses, a role for cytomegalovirus (CMV) (a member of the herpes virus family) in the onset and/or progression of AMD has been suggested. A post-mortem study showed that the posterior part of the eye could be a relatively common site of CMV latency in the general population. Furthermore, according to some in vitro and animal studies, CMV may participate in the onset/progression of AMD via pro-inflammatory and/or pro-angiogenic mechanisms.
Nevertheless, the impact of CMV on AMD remains largely unstudied in humans: only one small cross-sectional study has explored this association, showing a link between high levels of anti-CMV antibodies and neovascular AMD. However, determining whether CMV contributes to AMD onset or progression of AMD is essential, given the potential implications for treatment and prevention (antiviral therapies).
Using data from over 1,000 UK Biobank participants, we aim to assess whether CMV infection (estimated via anti-CMV plasma serology data) is associated with retinal layers thicknesses (measured by optical coherence tomography (OCT)). A particular focus will be given to the photoreceptor layer and the retinal pigment epithelium-Bruch’s membrane complex, which are known to be affected in AMD. Multivariate linear regression models will be performed to account for potential confounding factors. Moreover, thanks to the availability of genetic data in a subsample of participants, we will conduct stratified analyses on a genetic risk score for AMD to assess whether genetic background could modulate the impact of the virus on the risk of AMD.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/daily-patterns-of-physical-activity-and-syndromes-of-ageing-relationship-between-physical-inactivity-and-markers-of-geriatric-syndromes

Daily patterns of physical activity and syndromes of ageing. Relationship between physical inactivity and markers of geriatric syndromes.

Last updated:
ID:
33400
Start date:
23 November 2018
Project status:
Closed
Principal investigator:
Dr Sebastien Chastin
Lead institution:
Glasgow Caledonian University, Great Britain

GERIATRIC SYNDROMES are common clinical conditions prevalent in older people that do not fit into discrete disease categories. They include conditions such as falls, frailty, immobility, physical and cognitive decline; their impact on quality of life and disability is substantial. Although each one of these syndromes is a distinct health condition, geriatric syndromes share many common features (e.g. they are common in older populations especially the frail elderly). Although we still not fully understand why exactly they occur we believe that their origin is probably multi factorial. The aim of this study is investigate the relationship between daily patterns of (in) activity (a possible lifestyle determinant of tehe syndromes) and geriatric syndromes using the wealth of data available in the Biobank database. This 12 month project aims to improve our understanding of the relationship between patterns of daily physical activity and geriatric syndromes and to investigate whether they can provide a diagnostic of early signs of geriatric syndromes or be used as a preventive modality.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/dart-the-diet-and-arthritis-data-project

D+ART: The diet and arthritis data project

Last updated:
ID:
33557
Start date:
26 April 2019
Project status:
Closed
Principal investigator:
Dr Alex MacGregor
Lead institution:
University of East Anglia, Great Britain

Improving the quality of research into the role of diet in arthritis is a priority for patients. The aim of this application is to investigate dietary influences on three common musculoskeletal conditions: osteoarthritis, rheumatoid arthritis and chronic musculoskeletal pain. The analysis will focus on three specific aspects of diet for which there are commonly held beliefs about their potential for prevention and disease modification: the Mediterranean diet, diets high in fruit and vegetables, and diets low in fat. We will address the evidence for the claims about the benefits of these diets in the UK population.
The project will help provide consistent messages about the relationship between individual dietary exposures and arthritis to be communicated to patients. We will use these findings to inform strategies for prevention and treatment of these common musculoskeletal conditions, consistent with the aims of the biobank project.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/data-driven-based-correlation-and-prediction-analysis-of-ocular-surface-diseases

Data driven based correlation and prediction analysis of ocular surface diseases

Last updated:
ID:
98819
Start date:
3 March 2023
Project status:
Current
Principal investigator:
Dr Weijing Cheng
Lead institution:
Sun Yat-Sen University, China

Aims: The overall goal of this project is to analyze the crucial determinants and assess risk factors causally related to different kinds of ocular surface diseases, and to explore factors affecting corneal biomechanics. In addition to that, a deep learning system to predict the probability of occurrence and the corneal biomechanical alterations in these diseases will be built.
Ocular surface diseases, especially keratopathy and dry eye, are very serious challenges that bring enormous burden to patients and societies. Because no significant symptom can be observed at the early stages, many diseases may not be discernible until advanced stage, resulting in the impairment in visual functioning and vision-related quality of life. Along with the rapid development of deep learning these years, many deep learning models have been established in many ocular diseases. However, these models most focus on fundus lesions, we wonder whether ocular surface diseases could be predicted by using risk factors. Therefore, we want to use these data from different dimensions to explore the risk factors causally related to diseases, and finally build deep learning models to predict corneal biomechanical alterations and ocular surface diseases. Also, we need to validate these models.
If these models are successfully developed, many bothered by these diseases or at high risk of developing ocular surface lesions will fundamentally benefit from it, reducing medical cost.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/data-driven-decision-making-genetically-tailored-dietary-recommendation-for-preventing-metabolic-and-digestive-disorders

Data-driven decision making: Genetically tailored dietary recommendation for preventing metabolic and digestive disorders

Last updated:
ID:
78814
Start date:
6 December 2022
Project status:
Current
Principal investigator:
Dr Gengjie Jia
Lead institution:
Chinese Academy of Agricultural Sciences, China

With the concern of increasing incidence of metabolic and digestive disorders caused by unhealthy diet, this study aims to (1) identify risk factors that affect pathogenesis, (2) build mathematical models that can explain the processes involved in (1), and (3) develop dietary recommendation models/systems based on the models built in (2).

Using computational tools like machine learning, Mendelian randomization, and other statistical analyses, we can find out which genetic, environmental, and dietary factors play important roles in or associate with disease onset. Focusing on a few identified key factors, we will then investigate whether and how these factors work together in the process of disease pathogenesis. Based on all these findings, we will try to develop clinical biomarkers and build dietary recommendation models/systems tailored to the different needs of patients.

We anticipate that the project will take three years to accomplish above goals. Data pre-processing and series of association studies with genetics and environmental factors will be conducted in the first year. Then, finding the pattern with different data-driven approaches and retrospective cohort studies will be performed later on.

This research shall advance our understandings about the interplay of genetic and environmental factors that could explain the pathogenesis of metabolic and digestive disorders. We will develop an open web-based tool to differentiate the patients with metabolic and digestive disorders and then to recommend different diet patterns accordingly.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/data-driven-fast-and-high-quality-mri

Data-driven fast and high-quality MRI

Last updated:
ID:
100203
Start date:
21 November 2023
Project status:
Current
Principal investigator:
Mr Yinzhe Wu
Lead institution:
Imperial College London, Great Britain

Project Aim: The goal of this project is to reduce the time required for magnetic resonance imaging (MRI) scans while also improving the image quality. The goal is to make the process more efficient and pleasant for patients while retaining key clinical imaging features.

Scientific Importance: The recent advancement of artificial intelligence (AI) in medical imaging is being applied to a variety of clinical data, including MRI scans. The project’s goal is to use this technology to create clinically deployable tools for super-resolution (improving spatial resolution) and reconstruction (creating viewable images), which will improve the accuracy and reliability of the results.

Project Duration: 36 months

Public Health Benefits: The project will take 36 months to complete and will involve further improving the methodology as well as computational and clinical validation of the results. The hope is that this work will boost clinical team confidence in the images generated, resulting in more widespread use of these tools in standard clinical practise. This project is expected to improve patient care by reducing scanning time and allowing for greater access to MRI scans. Imaging quality advancements will also reduce artefacts and provide more detailed information for clinical interpretation, resulting in a tangible impact on the public healthcare system.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/data-driven-identification-of-a-drugs-disease-modifying-potential-outside-its-main-indication-area

Data-driven identification of a drug’s disease-modifying potential outside its main indication area

Last updated:
ID:
54717
Start date:
15 January 2020
Project status:
Closed
Principal investigator:
Professor Michael Krauthammer
Lead institution:
University of Zurich, Switzerland

The main aim of this study is the analysis of UK Biobank data to identify new uses for existing drugs. This may help patients without sensible drug options to get drug treatment without the need for lengthy and costly drug development. In this manner, a drug may potentially be repurposed for other uses.

When drugs are used for treating a specific disease or symptom, patients may experience additional unintended drug effects, referred to as drug side effects, which could be minor, such as a skin rash or more severe, such as gastrointestinal bleeding. In some cases, those effects might be considered beneficial and exploited for treatment. An example is a drug that is unexpectedly observed to cause low blood sugar. Interestingly, in patients suffering from diabetes, such a drug may actually lead to improvement of the disease.

To achieve our aim, we will proceed und use UK Biobank data as follows: based on the medications that patients have taken over time, and their medical history, we are trying to better understand additional unexpected consequences of those medications. We will then confirm these consequences by looking deeper at a person’s genomic profile, and associated clinical manifestations and blood values. Our final goal ist to generate a list of existing drugs and link them to potentially novel clinical applications. An additional outcome of our study is that physicians might be in a better position to understand the consequences of drug prescriptions and thus make more informative decisions for the treatment and care of their patients. In the following 24 months, we will use UK Biobank data for both establishing and validating the list of potential new drug uses, using the described analytical approaches.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/data-driven-identification-of-therapeutic-targets-for-non-invasive-brain-stimulation-in-mental-health-disorders-and-chronic-pain

Data driven identification of therapeutic targets for non-invasive brain stimulation in mental health disorders and chronic pain.

Last updated:
ID:
66061
Start date:
1 March 2021
Project status:
Closed
Principal investigator:
Dr Neil Bailey
Lead institution:
Monash University, Australia

Mental health conditions and chronic pain are highly prevalent, result in significant morbidity and mortality, and by and large lack treatments which are widely and highly effective. Development of novel approaches to treatment are urgently needed. Non-invasive brain stimulation approaches hold significant potential in the treatment of these disorders. Brain stimulation uses electricity to modulate neuronal firing patterns, this can be achieved in a number of ways including via direct electrical stimulation through electrodes placed on the scalp (transcranial Electrical Stimulation [tES]) or by inducing an electrical current in the brain via the application of highly focussed magnetic fields to the head (Transcranial Magnetic Stimulation [TMS]). Brain stimulation, namely TMS, is a well-established treatment for depression and has shown promising effects across a number of other mental health conditions. However, the effects of brain stimulation could be greatly improved through a better understanding of what type of brain activity in which brain regions to target in order to improve people’s symptoms.
Therefore, the aim of this research is to use a data driven approach to identify targets for brain stimulation treatment in a number of mental health conditions as well as chronic pain. We will do this through the use of sophisticated data analysis of comprehensive imaging and behavioural data.
Using this type of data driven approach to identify therapuetic targets will greatly enhance the progress of non-invasive brain stimulation treatment development across mental health conditions and chronic pain. The development of effective treatments for these conditions would have a significant and global positive impact.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/data-driven-stratification-of-chronic-pain-to-elucidate-the-contribution-of-inflammatory-mechanisms

Data-driven stratification of chronic pain to elucidate the contribution of inflammatory mechanisms

Last updated:
ID:
379089
Start date:
4 May 2025
Project status:
Current
Principal investigator:
Dr Yukiko Iwasaki
Lead institution:
Saitama Medical University, Japan

Aims: This research aims to uncover the biological mechanisms behind chronic pain by using advanced information sciences, such as medical informatics and omics technologies. We will analyze clinical big data to identify biologically similar groups among chronic pain patients.
Scientific Rationale: Chronic pain can be classified into three main types: psychosocial factors, nociceptive sensitivity, and neuropathic pain. These categories reflect various physiological and psychological factors involved in pain perception and regulation. However, they often overlap and intertwine in many patients, making the condition complex and challenging to treat.
The complexity of chronic pain’s underlying biology and the challenge of creating uniform study conditions for humans make this a difficult research area. Our study aims to address these challenges by leveraging clinical big data and advanced technologies to better understand and categorize chronic pain.
Methods: We will trace back records over the past three years to identify patients with diagnoses including pain. We will collect electronic health record information such as age, gender, diagnoses, medication history, test results, numerical pain scores, spreading pain sites, and treatment outcomes for this group of patients. Initially, we will conduct similarity analysis using a distributional semantic model to identify patterns of co-existing diagnoses among pain patients and predict underlying medical conditions for each.
For clinical information beyond diagnoses, such as blood test results, medication history, and pain scores, we will employ analytical methods such as hierarchical clustering in addition to traditional biostatistical techniques to stratify the patient population. We will investigate the relationship between classification based on diagnosis similarity and the characteristics derived from clinical information.
We will conduct comparative analysis to identify distinctive clinical information that characterizes each group. Based on the clinical information characterizing the generated patient groups, we will analyze underlying medical conditions and select groups where contributions to conditions such as chronic inflammation or metabolic changes are suspected to be significant. We will then validate these results using our clinical data from the Palliative Care Department at Saitama Medical University.
Project Duration: This project will be carried out over three years.
Public Health Impact: This research has the potential to significantly improve our understanding of chronic pain, leading to better diagnostic and treatment strategies. By identifying specific biological mechanisms and patient subgroups, we can develop more targeted and effective treatments, ultimately improving the quality of life for millions of people suffering from chronic pain.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/data-integrative-approaches-to-comorbidity-in-mental-disorders

Data-integrative approaches to comorbidity in mental disorders

Last updated:
ID:
44858
Start date:
26 November 2018
Project status:
Current
Principal investigator:
Professor Thomas Werge
Lead institution:
Institute for Biological Psychiatry, Denmark

One challenge in studying the causes of psychiatric disorders is that the disorders themselves are difficult to define. Patients may present varying, overlapping and evolving symptom profiles, making it difficult to pinpoint the nature of the disease and limiting the effectiveness of treatments. Hidden in the complexity, however, are immense opportunities. It has long been noted that secondary features, comorbid disorders and varying life outcomes, may define important subgroups of patients within and across traditional diagnostic boundaries. And such subgroups may display much more homogeneity, both in etiology and in response to medicine.

However, identifying and studying these subgroups requires large patient cohorts with extensive and diverse behavioral, disease, and sociodemographic measures that may not at first glance seem intuitively related to psychiatric outcomes. This important and unique advantage of the UK Biobank will allow us to use next generation data-integrative analytics to compliment traditional psychiatric diagnoses with an unprecedented wealth of secondary information. Over a three-year period, we aim to develop and apply new analytic approaches to better identify groups of patients that may share specific identifiable causes for their outcomes. The promise of the research is that it can provide a better framework for understanding the specific causes of mental disorders, be they genetic, environmental or evolutionary, and improve our ability to predict outcomes and target interventions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/data-mining-and-machine-learning-approaches-for-the-identification-of-interaction-partners-of-cholesterol-biosynthesis-intermediates-in-physiology-and-pathology

Data mining and machine learning approaches for the identification of interaction partners of cholesterol biosynthesis intermediates in physiology and pathology

Last updated:
ID:
618011
Start date:
25 July 2025
Project status:
Current
Principal investigator:
Mr Riccardo Perrone
Lead institution:
University of Parma, Italy

Cholesterol biosynthesis is a very complex and highly regulated metabolic process, in which the final product is well-characterized, but the metabolic intermediates are less so.
Focusing on the last ten distal reactions of cholesterol biosynthesis, which we can classify in Bloch and Kandutsch-Russell pathways, it has been seen that these enzymes and their metabolites (CBIs) play a fundamental role in human physiology and that their perturbations lead to pathological effects, such as Mendelian syndromes caused by inborn error of cholesterol biosynthesis.
Research suggests that several genetic variations in these enzymes are linked to an increased risk of neurodegenerative diseases, while their overexpression has also been observed in certain types of cancer.
This suggests that the sterol intermediates of cholesterol biosynthesis do not only act as precursors of cholesterol but that they are also involved in other physiological roles, beyond cholesterol production.
In this study we will associate rare, coding and non-coding genetic variants in genes of CBIs with clinical and molecular phenotypes, to understand in which pathologies they are implicated in, and we will perform combinatorial genetics analysis to determine the putative signaling mediators of these intermediates.
Despite the extensive knowledge about cholesterol, limited information is available on the molecular functions its distal intermediates. Considering how crucial cholesterol synthesis is, and that several widespread drugs target it, it is imperative to deeply understand the roles of CBIs outside of their function as biochemical precursors of cholesterol. The aim of this project is to improve the knowledge about CBIs and CBI genes through analysis on a large database such as UKBioBank.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/data-mining-for-cardiovascular-and-cerebrovascular-diseases-diagnosis-discrimination-in-the-uk-biobank

Data mining for cardiovascular and cerebrovascular diseases diagnosis discrimination in the UK Biobank

Last updated:
ID:
55917
Start date:
27 April 2021
Project status:
Current
Principal investigator:
Dr Alexandre Vallée
Lead institution:
Hôpital Foch, France

Cardiovascular and cerebrovascular diseases are major causes of mortality and are worldwide public health problem. Several factors, such as aging, hypertension, diabetes or dyslipidemia are associated with these diseases but are not specific.
Currently, no single correct diagnosis approach exits for patients in cardiovascular and cerebrovascular diseases prediction due to the variability in different clinical symptoms of patients and imperfection of diagnosis from noninvasive and invasive tests.
The objectives of our work were to use intelligence artificial to improve the accuracy decision diagnosis of cardiovascular and cerebrovascular diseases using all potential related risk factors. Data mining models could be interesting tools for prediction of Cardiovascular and cerebrovascular diseases and could participate to the detection of performed discrimination of cardiovascular and cerebrovascular diseases diagnosis. Data mining focuses machine learning, statistical analysis and databank technology. It assists the medical practitioner and analyst to mark intelligent medical decision which outmoded support system cannot.
The in-depth analysis of cardiovascular risk data will be spread over a three-year period to presenting several interesting results, particularly in Cardiovascular and cerebrovascular diseases.
Applied software production could be an interesting tool for cardiovascular and cerebrovascular diseases prediction. These models could be utilized such as a predictive tool in a personalized medicine in cardiovascular and cerebrovascular diseases diagnosis and prediction. Many studies will be needed to better evaluate these models and then estimate their acceptance by clinicians.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/data-science-approaches-to-the-early-diagnosis-of-human-diseases

Data science approaches to the early diagnosis of human diseases

Last updated:
ID:
116339
Start date:
30 January 2024
Project status:
Current
Principal investigator:
Professor Peter Duncan Donnelly
Lead institution:
University of St Andrews, Great Britain

We want to improve the early detection of diseases such as heart conditions, mental health disorders, and cancer. We want to understand why diseases occur together, which is known as multimorbidity. We are a cross-disciplinary group of scientists based at the University of St Andrews working in different fields like molecular biology, biophysics, bioinformatics, clinical science, and data science. Detecting diseases early is important because it helps doctors provide better treatments and gives patients a higher chance of recovery.
In this study, we propose to use a large database called the UK Biobank and take advantage of its multiple layers of data. These include genetic information, brain and body images, proteomic data, and information about people health in about 500,000 participants. We will use this resource to answer research questions generated in smaller scale study. By exploring the UK Biobank, we aim to discover new patterns and clues that can help us detect diseases earlier.
To achieve this, we will use different types of analysis. These include methods that allows scanning millions of genetic variants to identify genetic risk to diseases. We will also use machine learning approaches that can extract information from highly complex and large data sets that allow us to make predictions.
These advanced computer techniques will help us to develop models that can predict diseases at an early stage and identify people who are at a higher risk. Our goal is to gain a better understanding of diseases, improve early detection methods, and develop personalized treatments. The breadth and size of UK Biobank is a unique resource to allow such type of studies.
By working together as a cross-disciplinary team we will bring together the different expertise required to understand the highly diverse datasets and to implement different methodologies. Our research has the potential to save lives by enabling earlier detection of diseases and enhancing personalized healthcare.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/data-standardization-and-characterization-analysis-with-clinical-use-cases-relating-multiple-data-types-and-fields

Data standardization and characterization analysis with clinical use cases relating multiple data types and fields

Last updated:
ID:
119708
Start date:
22 January 2024
Project status:
Current
Principal investigator:
Mr Craig Mayer
Lead institution:
National Library of Medicine, United States of America

The aim of this project is to convert the format of the UK Biobank data to a commonly used format in order to use preexisting and reproducible methods to understand the dataset. We will than look at key information about the overall data and the population included in order to assess how the data is used and understand clinical factors about the population. With this key information from the converted form we will be able to compare the characteristics of the data to other similar datasets to find any similarities or differences that may exist. We will than look at individual conditions and other clinical characteristics to see how segments of the population compare to other datasets as well as look at how the converted data can be used to see results of clinical events and factors. All of this is done to help understand the data, how it can be used and how methods used on the data can be reused in other contexts for other datasets.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deciphering-amd-by-deep-phenotyping-and-machine-learning-pinnacle

Deciphering AMD by deep phenotyping and machine learning- PINNACLE

Last updated:
ID:
45477
Start date:
29 May 2019
Project status:
Closed
Principal investigator:
Professor Hrvoje Bogunovic
Lead institution:
Medical University of Vienna, Austria

Age-related-macular-degeneration (AMD) is a very common cause of blindness. Unfortunately, doctors don’t know who will progress to the sight threatening stage of the disease. Some patients progress slowly or not at all and others quickly.
We can teach computers to analyse high resolution images of the inside of the eye. We have access to hundreds of thousands of such images from patients with AMD and patients who don’t have AMD. These images together with those from UK Biobank will form a training data set, allowing us to train computers to identify what eye changes appear in patients with AMD. Once the computers have learnt this, we expect they will identify new changes we haven’t thought of.
Using this approach we think we will be able to better predict which patients will progress. This should help us develop better treatments and enter the most appropriate patients into clinical trials. It should allow us to better understand why AMD develops too.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deciphering-brain-diseases-deep-learning-analysis-of-brain-structure-and-disease-progression

Deciphering Brain Diseases: Deep Learning Analysis of Brain Structure and Disease Progression

Last updated:
ID:
135105
Start date:
13 March 2024
Project status:
Current
Principal investigator:
Professor Kyong Hwan Jin
Lead institution:
Korea University, Korea (South)

The expected value of this research lies in its potential to yield critical insights into brain diseases, facilitating early detection and intervention, ultimately enhancing public health. By employing cutting-edge deep learning techniques to analyze neuroimaging data, this study can uncover hidden disease patterns. This knowledge can translate into improved diagnostic methods and personalized treatment strategies, benefitting the wider public. Furthermore, the research contributes to the scientific community’s understanding of brain diseases, potentially reshaping healthcare approaches. In addressing a matter of significant public interest, this project aligns with the broader goal of enhancing brain health, thereby improving the quality of life for individuals affected by neurological conditions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deciphering-chronic-disease-association-studies-of-genome-exposome-and-phenome

Deciphering chronic disease – association studies of genome, exposome and phenome

Last updated:
ID:
92718
Start date:
25 August 2022
Project status:
Current
Principal investigator:
Dr Yanfeng Jiang
Lead institution:
Fudan University, China

Most chronic disorders are classified as multifactorial diseases (cardiovascular and cerebrovascular diseases, neurodegenerative diseases, metabolic diseases, and cancer, etc.). Better characterizing the interactions between environmental and genetic factors and investigating relationships between health-related outcomes is an important issue in understanding the biological mechanisms underlying these diseases. In this study, we aim to identify causal variants underlying disease risk, assess novel genetic associations across a wide spectrum of phenotypes, evaluate the phenotypic associations and genetic associations between chronic diseases, determine critical paths of disease connectivity through disease trajectory analysis, and identify novel biomarkers (derived from body fluids and imaging) linking to the genotype and phenotype. Our findings may have the potential to explore the pathogenic mechanisms of chronic diseases, identify biomarkers for early detection and possible targets for intervention, and provide solutions for precision medicine. The rolling period of the proposed program is three years, yet it might be prolonged due to methodological updates and novel findings which need to be further validated. Our objectives are aligned with UK Biobank’s goal to improve the prevention, diagnosis, and treatment of a wide range of serious and life-threatening illnesses.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deciphering-complex-traits-phenotypic-and-genetic-associations-between-traits-in-the-uk-biobank-cohort

Deciphering complex traits – phenotypic and genetic associations between traits in the UK Biobank Cohort

Last updated:
ID:
54803
Start date:
29 October 2019
Project status:
Current
Principal investigator:
Dr Huan Song
Lead institution:
Sichuan University, China

Most diseases are categorized as complex. These diseases or health-related outcomes result from a complex interplay of genes and environment/lifestyle factors. Investigating relationships between health-related outcomes can provide a unique opportunity to unravel common causes of complex diseases and shed light on the biological mechanisms underlying these diseases. With particular interests on complex diseases that confer a major impact on public health, we will primarily focus on mental disorders, cancer, cardiovascular diseases, infections and autoimmune diseases, and aging-related diseases.

Specifically, we aim to identify genetic markers of complex diseases, assess the phenotypic associations between complex diseases, evaluate the genetic associations across complex diseases, and identify brain imaging markers linking genetic makeup to mental symptoms/disorders. Our findings may have the potential to significantly advance our knowledge of the biological basis for health-related complex diseases, leading to refined disease prevention and novel markers for early detection and/or intervention. The proposed program will last for three years but it might be prolonged due to advances in methodology and novel findings which may require external validations.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deciphering-extraordinary-autobiographical-memory

Deciphering extraordinary autobiographical memory.

Last updated:
ID:
101531
Start date:
28 October 2024
Project status:
Current
Principal investigator:
Dr David Coynel
Lead institution:
University of Basel, Switzerland

Everyone remembers a certain number of significant events that happened in his or her own life. These events usually have an emotional component, such as the birth of a child or the death of a relative. But remembering what you had for breakfast on a specific day 6 years ago is not relevant and you most certainly don’t remember it. There are however some people who do remember such events. They can actually remember every day of their life, dating back to mid-childhood. These individuals are not extraordinary learners, with average memory performance on other memory tasks. The exact mechanism behind this atypical ability is unknown. We have the unique opportunity to acquire high resolution brain images of one such individual, and investigate whether certain regions or connections in his brain also are out of the norm. To properly answer such a question, we need a large and carefully matched set of control male participants in the same age range, with identical brain imaging information. Only the UK Biobank can provide us with such a population, together with the tools to analyse the data in a similar manner. Understanding such extraordinary capacities could in the long run prove useful to better understand memory deficits occurring with age and psychiatric diseases. We hope to conduct the project to its end in 2023.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deciphering-genetic-association-between-cancer-incidence-and-cardiovascular-disease

Deciphering genetic association between cancer incidence and cardiovascular disease.

Last updated:
ID:
407461
Start date:
5 November 2024
Project status:
Current
Principal investigator:
Professor Sangwoo Kim
Lead institution:
Yonsei University, Korea (South)

This study aims to explore the genetic links between heart disease, specifically cardiomyopathy, and cancer. We will examine genetic mutations in cardiomyopathy-related genes to see how they might contribute to both conditions. By comparing the genetic data of cancer patients and individuals without cancer, we hope to uncover important connections that could lead to better prevention and treatment strategies for these diseases.

Its findings could significantly impact public health by improving early detection and developing more effective treatments for people at risk of both heart disease and cancer.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deciphering-genetic-variations-and-clinical-phenotypes-through-large-scale-integrated-analysis-of-the-uk-biobank-cohort

Deciphering Genetic Variations and Clinical Phenotypes through Large-Scale Integrated Analysis of the UK Biobank Cohort.

Last updated:
ID:
1023249
Start date:
30 October 2025
Project status:
Current
Principal investigator:
Dr Dae-Soo Kim
Lead institution:
Korea Research Institute of Bioscience and Biotechnology, Korea (South)

This project aims to leverage the extensive genomic and clinical resources of the UK Biobank cohort to systematically explore the relationships between genetic variations and diverse clinical phenotypes. The central research questions are:
(1) Which genetic variants are significantly associated with disease onset, progression, and prognosis? (2) How do pleiotropic variants influence multiple phenotypic traits across different disease domains? (3) Can integrated genomic and clinical data improve the accuracy of risk prediction models beyond conventional approaches?
The objectives of this study are threefold. First, to conduct comprehensive genome-wide and phenome-wide association studies (GWAS and PheWAS) to identify genetic determinants of complex traits and diseases. Second, to integrate genomic data with clinical, lifestyle, and environmental information, thereby uncovering gene-environment interactions and pleiotropic effects. Third, to employ advanced machine learning and artificial intelligence methods to develop predictive models for disease risk, stratification, and prognosis, facilitating translation into precision medicine applications.
The scientific rationale is grounded in the unprecedented scale and depth of the UK Biobank dataset, which offers both statistical power and diversity to detect novel associations. Traditional single-omics or genotype-only studies are limited in their ability to capture the complexity of human health and disease. By unifying genomic, clinical, and environmental data, this project will provide deeper biological insights into disease mechanisms, reveal novel therapeutic targets, and establish robust predictive frameworks.
Ultimately, the findings are expected to contribute to early disease detection, risk stratification, and personalized interventions, thereby advancing precision medicine and improving healthcare outcomes on a population-wide scale.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deciphering-genome-exposome-phenome-interplay-underlying-neuropsychiatric-disorders

Deciphering Genome-Exposome-Phenome interplay underlying neuropsychiatric disorders

Last updated:
ID:
432214
Start date:
6 December 2024
Project status:
Current
Principal investigator:
Professor Deng-Feng Zhang
Lead institution:
Kunming Institute of Zoology, China

The molecular mechanism of complex diseases, like Alzheimer’s disease, remains unclear regarding numerous genetic and environmental risk factors, since each contributes to the disease with a tiny effect. The detection of interactions between risk genes, and between genotypes (e.g. APOE4) and risk factors (e.g. HSV-1 infection history), requires longitudinal cohort with deep phenotyping and genotyping. Using the UK Biobank cohort with genome, exposome, and deep phenotype information, this project aims to identify the risk factors underlying typical neuropsychiatric disorders, and explore the interaction between the genetic and environmental factors. The project will last for about 36 months, with comprehensive data analyses and experimental validations of the findings. The research will provide detailed understanding of the mechanism of complex neuropsychiatric diseases, making a complement to the traditional study which mainly focused on single risk factor. Such knowledge may provide potential targets for the precise medicine of these complex disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deciphering-inflammaging-unraveling-the-interplay-of-inflammation-aging-and-neurodegeneration-in-parkinsons-disease

Deciphering Inflammaging: Unraveling the Interplay of Inflammation, Aging, and Neurodegeneration in Parkinson’s Disease.

Last updated:
ID:
200895
Start date:
3 October 2024
Project status:
Current
Principal investigator:
Miss Yuchen Yan
Lead institution:
Fudan University, China

Our research project explores the connection between the immune system’s changes with aging and the development of neurodegenerative diseases like Parkinson’s Disease (PD). The concept of “inflammaging” describes a state of chronic, low-grade inflammation associated with aging, believed to contribute to the progression of age-related diseases, including PD.

**Aims:**
– To investigate the link between inflammaging and neurodegenerative diseases.
– To identify blood and bodily fluid markers that could signal the risk of PD.
– To assess if immune-modifying treatments could lower PD risk.

**Scientific Rationale:**
As we age, our immune system can begin to function improperly, sometimes attacking our own cells or causing harmful inflammation. This misdirected immune activity is suspected to be a key factor in many neurodegenerative diseases. Understanding these processes could reveal new prevention or treatment strategies.

**Project Duration:**
The study will span several years, utilizing data from the UK Biobank, which includes health information from half a million UK residents. This comprehensive analysis aims to dissect the relationship between the immune system, aging, and neurodegeneration in detail.

**Public Health Impact:**
Neurodegenerative diseases significantly impact individuals’ lives and healthcare systems. Our findings could lead to new ways to predict, prevent, or slow these diseases, improving quality of life and reducing healthcare burdens. Identifying early disease risk markers linked to the immune system and aging could help develop strategies for better brain health in older age, benefiting millions worldwide.

In essence, our project aims to elucidate how aging and the immune system interact to affect brain health. Insights from our research could open up new avenues for preventing neurodegenerative diseases, promoting healthier aging populations.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deciphering-of-the-casual-genetic-relationships-between-rheumatoid-arthritis-and-varying-comorbidities

Deciphering of the casual genetic relationships between rheumatoid arthritis and varying comorbidities

Last updated:
ID:
85194
Start date:
25 August 2022
Project status:
Current
Principal investigator:
Professor Gregory Livshits
Lead institution:
Tel Aviv University, Israel

Rheumatoid arthritis (RA), a debilitating autoimmune condition is associated with several accompanying ailments. RA and its accompanying diseases appear to have genetic components. We aim to find the shared genetic architecture involved between RA and its shared diseases, so that we can hopefully clarify the shared pathway involved in the development of RA and its shared diseases. The future of medicine is precision medicine, and by clarifying the genetic profile of RA, we hope to advance therapeutic targets that may aid in treating or targeting RA development.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deciphering-recurrent-surgical-ent-diseases-through-genetics-and-deep-phenotyping

Deciphering recurrent surgical ENT diseases through genetics and deep phenotyping

Last updated:
ID:
117308
Start date:
28 March 2024
Project status:
Current
Principal investigator:
Ioannis Vlastos
Lead institution:
Evaggelismos Hospital, Greece

Our research project focuses on two prevalent surgical ENT diseases – cholesteatoma and laryngeal cancer – and aims to tackle their high recurrence rates, which contribute to their significant impact on patients’ health. By combining genetic studies with clinical and laboratory data from a large number of patients, we hope to make a difference in how these conditions are treated.
For cholesteatoma cases, we will harness the power of Artificial Intelligence (AI) to explore potential complementary medical treatments. By analyzing a vast amount of data from the UK Biobank, we aim to identify new therapeutic options that could help reduce the chances of recurrence.
In the case of laryngeal cancer, our goal is to create a nomogram – a predictive tool – that can guide healthcare professionals in choosing the most effective treatment for each patient (chemoradiation vs laryngectomy).
Our project will last for one year, during which we will analyze the extensive data available in the UK Biobank to develop predictive models for both cholesteatoma and laryngeal cancer. To ensure the reliability of our findings, we will validate these models using data from patients treated at our hospital, representing different nationalities.
The impact of our research could be profound. By understanding the genetic factors underlying recurrences in these ENT diseases, we aim to pave the way for improved treatment strategies. Finding complementary medical treatments for cholesteatoma and optimizing treatment choices for laryngeal cancer could lead to better patient outcomes and reduced morbidity rates. Ultimately, our project aspires to contribute positively to public health by offering more effective and personalized treatment options for individuals facing these challenging medical conditions


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deciphering-relevance-of-interplay-between-lipoproteins-and-lipid-metabolites-for-cardiovascular-disease-risk-with-genetic-clustering

Deciphering relevance of interplay between lipoproteins and lipid metabolites for cardiovascular disease risk with genetic clustering.

Last updated:
ID:
570811
Start date:
19 August 2025
Project status:
Current
Principal investigator:
Mr Jakub Morze
Lead institution:
Chalmers University of Technology, Sweden

Research Questions:
1. How do genetic determinants of lipid metabolites interact with lipoproteins to influence CVD risk?
2. Can genetic clustering of lipid-lipoprotein interactions provide novel insights into lipid-mediated disease mechanisms?
3. Do polygenic risk scores (PRS) derived from lipid-lipoprotein clusters predict intermediate atherosclerotic phenotypes and CVD outcomes?

Objectives:
1. Develop partitioned polygenic risk scores (PPRS) reflecting genetically determined lipid-lipoprotein interactions using genome-wide association study (GWAS) data.
2. Validate the generated PPRS against measured lipid metabolites in cohorts with available lipidomic data.
3. Assess the predictive value of these PPRS for incident CVD events in the UK Biobank (UKB).

Scientific Rationale:
Lipidomics studies have identified numerous lipid metabolites associated with CVD, yet their effects are often confounded by their lipoprotein carriers. Most lipidomics analyses measure total plasma lipid levels without considering the class-specific origin of these metabolites. Since different lipoproteins (e.g., LDL, VLDL, HDL) exhibit distinct lipid compositions and atherogenic properties, it is crucial to disentangle the role of lipoprotein-specific lipids in CVD risk.
Genetic clustering provides a powerful tool to define biologically relevant lipid-lipoprotein interactions. By leveraging UKB data, this project will apply machine-learning-based genetic clustering to classify variants associated with both lipid metabolites and conventional lipoprotein measures (LDL-C, triglycerides, Lp(a), HDL-C). The resulting genetic clusters will be used to construct PPRS, which will be tested for associations with imaging-based markers of atherosclerosis, metabolic dysfunction, and incident CVD events.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deciphering-the-biological-basis-of-and-identifying-therapeutic-targets-for-alzheimer-disease

Deciphering the biological basis of and identifying therapeutic targets for Alzheimer disease

Last updated:
ID:
101549
Start date:
30 November 2023
Project status:
Current
Principal investigator:
Professor Gyungah R Jun
Lead institution:
Boston University (US), United States of America

Alzheimer disease (AD) is a progressive neurodegenerative disease that is a staggering burden for those affected and their families, public health and society in general. Since 2003, 99% of drug trials for AD did not yield efficacious results and thus failed to get approval from the United States Food and Drug Administration (FDA). These disappointing outcomes support the idea that AD is multifactorial and etiologically heterogeneous disease. Therefore, there is an urgent need to develop multiple prevention and treatment options tailored to one’s risk profile based on genetic, biomarker and other measurable factors. The proposed project will investigate extensive genetic, biomarker, brain imaging, cognitive, environmental exposure and lifestyle data to identify genetic factors that are associated with AD and AD-related traits using UKBB and other sources of data. To accomplish the objective, we will (1) attempt to replicate genetic association findings established in external datasets using UKBB data; (2) analyze a wide array of genetic, genomic, laboratory-based, cognitive, brain imaging and other data in the UKBB using multiple statistical and computational approaches to identify disease pathways and establish AD risk profiles; (3) prioritize drug targets for each risk subgroup, and (4) investigate differential medication effects between low- and high-risk network subgroups. By evaluating relationships of risk profiles of UKBB participants with health data including medication usage and bioassay data, we will identify and prioritize novel as well as existing drug targets for drug development or repositioning, respectively. The proposed work will establish a robust foundation of fast-track clinical translation for drug targets and repurposing via clinical trials.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deciphering-the-biological-role-of-headache-and-migraine-genetic-risk-variants

Deciphering the biological role of headache and migraine genetic risk variants

Last updated:
ID:
71718
Start date:
21 September 2021
Project status:
Current
Principal investigator:
Dr Daniel Baksa
Lead institution:
Semmelweis University, Hungary

Headache disorders can negatively affect our life by causing sufferings and limiting our activities and ability to work. It is already known that genetic background, environmental factors, and other disorders can influence headaches and one of their most important members, migraine. Several drugs for treatment already exist in clinical practice, but the failure of these drugs are relatively common, especially in migraine. Although new drugs recently became available but these are also not effective for everyone probably because headaches and migraine can develop for several reasons. In our analysis, we approach the problem from a genetic perspective. Previous studies with animals and in different cell types (especially neurons) can help us to identify biological mechanisms with high importance in headaches. Among them some showed the opportunity of therapeutic application. In our studies, we plan to identify genetic variants that belong to these biological mechanisms and determine their relationship with headache and migraine in the UK Biobank dataset. We will use this information to create specific genetic risk scores that can characterise the biological mechanism we would like to investigate and use the genetic risk scores in other studies to determine how they influence brain function (measured by functional magnetic resonance imaging or shortly, fMRI) in healthy controls and headache patients. We will also investigate whether genetic risk scores or specific genetic variants influence the effect of the environment (e.g. income), other disorders or lifestyle (e.g. sport, diet) on the development of headaches. We believe that our complex approach can lead to a better understanding why headaches and migraine, in particular, are so diverse in their development and response to treatment, and may aid a more personalised treatment in headache patients in the future.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deciphering-the-complex-relationship-between-asthma-allergy-and-pancreatic-cancer-risk

Deciphering the complex relationship between asthma/allergy and pancreatic cancer risk

Last updated:
ID:
94757
Start date:
13 December 2023
Project status:
Current
Principal investigator:
Dr Nuria Malats
Lead institution:
National Cancer Research Center, Spain

Asthma and allergies are very common diseases in the population that begin during childhood. Suffering from asthma has been associated with a reduced risk of some cancers, including pancreatic cancer. This is one of the most aggressive tumors, which is increasing in frequency in our environment and for which there is no effective treatment because patients are diagnosed too late. Therefore, it is essential to diagnose pancreatic cancer at earlier stages. With this project we want to study whether asthma and allergies are causally associated with pancreatic cancer, as well as the role of other diseases, e.g. obesity, exposures such as smoking, and also whether the treatment of asthma and allergies affect this association. In addition, we will want to investigate how local inflammation in the pancreas and systemic inflammation combine to develop pancreatic cancer, or one of its subtypes. To dissect this puzzle, we will apply a unique combination of epidemiological methods, tools and resources, a large number of biomarkers, data science and advanced computational methods, as well as the proven expertise of researchers from different disciplines.
The identification of the asthma endotype that confers protection to the development of specific subtypes of pancreatic cancer will provide the clues to further investigate the cellular/molecular mechanisms associated with this type of asthma and to find immunological strategies to prevent pancreatic cancer. If the results show that drugs used in the treatment of asthma protect against the development of pancreatic cancer, their use could be considered for the prevention of this cancer in high-risk patients (e.g. patients with a family history or with late-diagnosed diabetes). If these drugs have a poor toxicity profile, it will be necessary to determine through which mechanisms they exert this action in order to identify others that may be more selective and can be used in the clinical setting. In case of identifying peripheral blood markers associated with a protective or risky immunological environment at the tumor level, it will be possible to use them to monitor patients with a patients with an increased risk of developing pancreatic cancer.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deciphering-the-genetics-of-cachexia

Deciphering the genetics of cachexia

Last updated:
ID:
17658
Start date:
13 June 2016
Project status:
Closed
Principal investigator:
Dr Jan Korbel
Lead institution:
EMBL Heidelberg, Germany

Cachexia and cancer-associated cachexia (CAC) are characterized by extensive body weight loss, loss of fat tissue (white adipose tissue), and skeletal muscle. In cancer patients, CAC is a positive risk factor for death. The molecular mechanisms underlying cachexia and CAC are currently only poorly understood, thus limiting therapeutic intervention. Our laboratory has evidence that cachexia and CAC have an underlying genetic component, and we propose to perform a genome-wide association study (GWAS) to uncover the genetic underpinnings of cachexia/CAC in the context of cancer, infectious and inflammatory disease with the help of genotype and phenotype data from the UK Biobank. Cachexia and especially cancer-associated cachexia (CAC) is characterized by extensive body weight loss, loss of fat tissue, and skeletal muscle, and other phenotypes including wide-spread inflammation. CAC occurs in approx. 50% of cancer patients, depending on its definition, and is an independent predictor for cancer-related death (PMID:21296615). We envision our project will yield novel insights into the biological mechanisms of cachexia in the context of cancer, infectious and inflammatory disease. Validation of our findings could pave the way for cachexia/CAC intervention strategies, and results of our health-related research study is thus of relevance for patients and the public in general. We aim to identify genetic markers associated with rapid weight loss in individuals diagnosed with cancer, infectious and inflammatory disease to obtain novel insights into cachexia and especially CAC. For this, we will perform an unbiased genome-wide association study to identify association between patients? genotype and weight loss. Weight changes will be determined using weight assessment at repeated timepoints and/or adipose tissue changes as determined by imaging. We propose to pursue our analyses with the full cohort currently available, limited to patients with cancer, infectious and inflammatory disease for which genotyping data are available. We will include in our analysis cancer types with an incidence of at least 50 across the UK Biobank cohort (including colorectal cancer and pancreatic cancer), inflammation-related diseases with an incidence of at least 50 across the UK Biobank cohort (including chronic kidney disease, cystic fibrosis, multiple sclerosis, and Rheumatoid arthritis), and infectious diseases (including tuberculosis, HIV) with an incidence of at least 50 across the UK Biobank cohort.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deciphering-the-genomic-architecture-of-three-major-cancers-in-african-ancestry-population

Deciphering the genomic architecture of three Major Cancers in African-Ancestry Population

Last updated:
ID:
1026225
Start date:
6 November 2025
Project status:
Current
Principal investigator:
Professor Olubanke Olujoke Ogunlana
Lead institution:
Covenant University, Nigeria

Background & rationale. Prostate, breast and colorectal cancers often present more aggressively in people of African ancestry, yet genome-wide association studies (GWAS) for these populations remain severely under-powered. Leveraging the self-identified Black/African UK Biobank participants-together with 1000 Genomes African reference panels-we will address this gap and improve equity in cancer genetics and possible improved precision medicine.
Research questions.
1. Which single-nucleotide polymorphisms (SNPs), genes and biological pathways underlie these three cancers in African-ancestry individuals?
2. How do risk loci, fine-mapped causal variants and polygenic risk scores (PRS) compare with findings in European-ancestry cohorts?
3. Can machine-learning (ML) models that incorporate African-specific effect sizes outperform conventional European-derived PRS in predicting cancer risk?
Objectives.
* Perform stringent QC, ancestry clustering and fine-scale sub-ancestry inference.
* Conduct single-variant GWAS with linear mixed models (LMM & EDLMM) and gene-based tests with SKAT.
* Integrate expression-weighted association tests (MOKA) and perform pathway enrichment (GO, KEGG, Cancer Mine).
* Apply LD-aware fine-mapping with MAGMA to prioritise likely causal variants.
* Build and benchmark ML classifiers and PRS (p < 0.05 filters) within African participants and compare performance to PRS trained in Europeans.
* Release summary statistics and code to advance cancer genetics in under-represented populations.
Impact. Identifying African-specific genetic risk factors will (i) improve our understanding of cancer aetiology, (ii) provide a foundation for ancestry-tailored screening and prevention strategies, and (iii) reduce disparities in genomic medicine.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deciphering-the-impact-of-built-and-atmospheric-environment-on-mental-and-cognitive-health-dementia-and-mortality-outcomes-utilising-the-ukbump-database-a-dpuk-supported-study

Deciphering the impact of built and atmospheric environment on mental and cognitive health, dementia and mortality outcomes utilising the UKBUMP database: a DPUK supported study

Last updated:
ID:
44469
Start date:
18 September 2019
Project status:
Current
Principal investigator:
Dr Chinmoy Sarkar
Lead institution:
University of Hong Kong, Hong Kong

The aim of this research project is to understand the associations between air pollution exposure and built enviroment on multiple physical, mental and cognitive health outcomes, dementia and mortality.

1. The proposed research will aim to understand the links from air pollution exposure to mental health, cognitive function (and dementia), chronic disease, and mortality, and how the relationship is configured by urban built environment, lifestyle, SES and genetic factors. Causal evidence generated from the study will inform strategies and policies towards minimising exposures from health inhibiting urban environments, especially harmful air pollutants.

2. We hypothesize that participants residing nearer to healthcare facilities have higher utilization of care services and hence lower mortality, reduced chronic disease prevalence and length of stays in hospitals. The proposed project aims to test this hypothesis among the UK Biobank participants (N=500,000) employing the healthcare accessibility metrics within the UK Biobank Urban Morphometrics Platform (UKBUMP).

The evidence thus generated will help optimize healthcare service allocation with an aim to minimize mortality, DALYs and healthcare expenditures. The evidence will also inform strategies for pollution management in major cities around the world.

The project length will be 36 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deciphering-the-impact-of-environmental-determinants-on-digestive-system-disease-and-other-chronic-diseases-based-on-the-multi-omic-data

Deciphering the impact of environmental determinants on digestive system disease and other chronic diseases based on the multi-omic data

Last updated:
ID:
232231
Start date:
19 August 2025
Project status:
Current
Principal investigator:
Dr Jie Chen
Lead institution:
Third Xiangya Hospital of Central South University, China

The prevalence of digestive system disease and chronic diseases is increasing. These diseases are often accompanied by chronic disabling conditions, which have significant socio-economic implications for individuals, healthcare systems, and society. A combination of genetics, environmental factors and lifestyle determine an individual’s risk of disease susceptibility. Advancing multi-omics technology allowing exploration of pathophysiological factors during the disease process, and the UK Biobank provides a wealth of information for us as a large dataset containing multi-omics, broad environmental exposure and clinical disease data. We propose this study to address a series of research questions in how a range of environmental risk factors are associated with digestive system diseases and chronic disease risk, prognosis and survival through multi-omics data. We explored the multi-omics signatures of digestive system and chronic diseases using genomics, proteomics, and metabolomics data. Obtaining disease-associated signature changes will help to elucidate the pathophysiological processes involved in disease development. We will map how common risk factors influence disease to gain inspiration for early prevention and clinical intervention. Our planned research aligns with the objectives of UK Biobank research, which aims to improve the prevention, diagnosis, and treatment of disease and promote the health of society as a whole. The findings of this work will provide solid evidence for the complex pathogenesis of digestive system and chronic diseases and for patient clinical practice.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deciphering-the-impact-of-structural-variation-on-human-genetic-diversity

Deciphering the impact of structural variation on human genetic diversity

Last updated:
ID:
78690
Start date:
15 November 2022
Project status:
Current
Principal investigator:
Dr Peter Heshedahl Sudmant
Lead institution:
University of California, Berkeley, United States of America

Any two humans share 99.9% of their DNA sequence. Differences between genomes are due to single nucleotide polymorphisms (SNPs) as well as changes in genome structure. These “structural variants” include duplications, deletions, and inversions of whole blocks of DNA. While a great deal of focus has been placed on analysis of SNPs, structural variants contribute more differences to human genomes than SNPs and are important contributors to human diversity and disease susceptibility.

One region of structural diversity is a large inversion on chromosome 17 referred to as the 17q21.31 inversion locus. This region of the genome has medical relevance and is implicated in disease – including Parkinson’s, tau-related dementia, and autism spectrum disorders as well as a very severe spontaneous disease called Koolen-de Vries syndrome. To better understand these diseases, we are looking at the inversion’s structure throughout the UK Biobank. The inversion is present in two different forms (called “haplotypes”) one of which is present in Europeans at a higher frequency than in other populations. Understanding the history of this locus not only provides a window into human demography but contributes to our understanding of the diseases associated with this region of the genome.

Several regions of the genome exhibit increased susceptibility to disease causing structural variants. Our work proposes to study these regions to identify how these regions evolve and how they might impact human health and disease. Over the next three years, we hope to systematically examine the UK Biobank dataset to analyze this diversity probing inversion loci (such as 17q21.31 ) and to additionally explore other structurally complex loci in the genome.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deciphering-the-metaboloc-and-immune-cross-talk-in-cancer-patients

Deciphering the metaboloc and immune cross-talk in cancer patients

Last updated:
ID:
218074
Start date:
26 November 2024
Project status:
Current
Principal investigator:
Dr Stefano Cacciatore
Lead institution:
International Centre for Genetic Engineering and Biotechnology, South Africa

During the last few years, we have put a concerted effort in characterising the metabolic and lipoprotein profile of prostate, pancreatic and liver cancer in with some African cohorts. In pancreatic and liver cancer, we identified a metabolite marker strongly associated with the survival time. We believe that this marker is associated with a high oxidative stress in the liver and consequently could have a strong impact in the immune system.
We aim to compare our results obtained in African patients with the data from the UK Biobank.
The impact of this study will directly benefit cancer patients since it may lead to the identification of non-invasive early prognostic biomarkers and the identification of metabolic pathways that can be targeted to counteract disease progression and therapeutic efficacy.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deciphering-the-role-of-emotional-distress-and-functional-disability-among-patients-with-chronic-musculoskeletal-pain

Deciphering the role of emotional distress and functional disability among patients with chronic musculoskeletal pain

Last updated:
ID:
80584
Start date:
12 January 2022
Project status:
Current
Principal investigator:
Professor Shiqing Feng
Lead institution:
Qilu Hospital of Shandong University, China

Based on the International Classification of Diseases 11th Revision, the definition of chronic pain (chronic musculoskeletal pain is one main category) should include significant emotional distress and/or significant functional disability. However, no further details to define the term “significant”. This study will assess the prognosis for patients with chronic musculoskeletal pain considering their emotional distress and functional disability, which might help us identify the high-risk population in advance. Moreover, it is still unclear whether there is an association (maybe bidirectional) between emotional distress and functional disability among patients with chronic musculoskeletal pain as previous studies were limited by methodological limitations (e.g. cross-sectional design and small sample size). This study will quantify the association between emotional distress and functional disability among patients with chronic musculoskeletal pain, which might provide new insights into understanding chronic musculoskeletal pain. Finally, it is challenging to manage patients with chronic musculoskeletal pain, especially considering their emotional distress and functional disability. Causal mediation analysis is a powerful tool to explore the potential mediation role of lifestyle behaviours (e.g. smoking, alcohol consumption and physical activity), pharmacological and non-pharmacological treatment. The results from causal mediation analysis might help us optimize current therapies and guide the development of new interventions. We plan to finish this project in 36 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deciphering-the-role-of-lifestyle-and-environmental-exposures-in-ovarian-aging

Deciphering the Role of Lifestyle and Environmental Exposures in Ovarian Aging

Last updated:
ID:
262263
Start date:
5 August 2025
Project status:
Current
Principal investigator:
Dr Guiquan Wang
Lead institution:
Xiamen University, China

Ovarian aging, a natural facet of biological aging, precedes the functional decline of other organ systems by approximately a decade. It is marked by a progressive decrease in the quantity and quality of oocytes, leading to a significant decline in ovarian function with profound implications for quality of life and well-being. Lifestyle factors, including diet, smoking, exercise, and stress levels, have been shown to influence the rate of ovarian aging, with diets rich in antioxidants potentially slowing this process. However, the complex interplay and cumulative effects of these factors on ovarian aging remain to be fully understood, as do the impacts of other lifestyle and environmental factors such as alcohol consumption, sleep patterns, social interactions, and environmental exposures.

Previous research has identified numerous genetic determinants of ovarian aging, including 290 genetic markers linked to the age at natural menopause in women of European ancestry and 195 pathogenic variants associated with primary ovarian insufficiency. Despite these advances, the connection between lifestyle factors and ovarian aging through gene expression, protein activity, or metabolic changes is underexplored. Bridging human genetics with large-scale proteomics and health outcome phenotypes could shed light on the influence of lifestyle and environmental factors on ovarian aging.

This proposal seeks to explore novel factors associated with ovarian aging by leveraging data from the UK Biobank to examine the relationship between lifestyle, environmental exposures, and ovarian aging over three years. It will include medical conditions related to ovarian aging, blood biomarkers, and physical measurements for comparison, integrating genetic and plasma proteomics data to construct a network of lifestyle factors, gene expression, and protein activities. By employing machine learning models, the project aims to predict how various lifestyle factors affect ovarian aging and identify key regulatory mechanisms.

This research has the potential to reveal new lifestyle predictors of ovarian aging, offering accessible insights for the public to potentially prolong reproductive health and inform public health strategies for reproductive health maintenance.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deciphering-the-temporal-trajectory-of-apoe-and-inflammation-related-to-metabolic-and-vascular-risk-factors-on-brain-integrity

Deciphering the Temporal Trajectory of APOE and Inflammation Related to Metabolic and Vascular Risk Factors on Brain Integrity

Last updated:
ID:
609424
Start date:
19 August 2025
Project status:
Current
Principal investigator:
Ms Yumiko Wiranto
Lead institution:
University of Kansas Medical Center, United States of America

The risk of developing Alzheimer’s disease (AD) has been associated with genetic and health factors, such as APOE4 and metabolic and vascular risk factors (MVRFs). Due to the irreversible nature of AD, early detection for intervention is critical. Magnetic resonance imaging (MRI) has been instrumental in identifying brain changes that precede clinical manifestations by several years. These changes have been observed in various MRI phenotypes, including brain volume, cortical thickness, white matter tracts, and white matter hyperintensities, as well as cognitive outcomes.
However, the findings on these phenotypes have shown some inconsistencies. For instance, some studies reported increased brain volume and cortical thickness in APOE4 carriers or individuals with MVRFs, while others noted cortical thinning and reduced volume. These discrepancies may stem from the hypothesized antagonistic pleiotropic effects of APOE4, or the early inflammatory response induced by MVRFs, contributing to temporal hypertrophy. Notably, these factors appear to impact men and women in a different manner or degree. Because MVRFs are linked to lifestyle behaviors, such as alcohol consumption, smoking, sleep, and physical activity, these factors will also be included in this project.
To address these complexities, we aim to conduct the following analyses separately for each sex: 1) analyze the temporal dynamics of APOE genotype and MVRFs with various MRI phenotypes, with inflammatory markers as mediators. 2) define the role of lifestyle behaviors on the relationship between MVRFs and various MRI phenotypes based on APOE genotype. 3) test how these factors collectively affect cognitive performance. We hypothesize that there will be sex differences in the effects of MVRFs and inflammation on the brain and cognition, and the effects will be more pronounced in APOE4 carriers.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/decoding-cardio-renal-crosstalk-in-coronary-artery-disease-using-uk-biobanks-multi-organ-imaging-and-multi-omics-data

Decoding Cardio-Renal Crosstalk in Coronary Artery Disease Using UK Biobank’s Multi-Organ Imaging and Multi-Omics Data

Last updated:
ID:
778385
Start date:
9 July 2025
Project status:
Current
Principal investigator:
Dr Boxuan Feng
Lead institution:
Tianjin University of Traditional Chinese Medicine, China

This 3-year project aims to address critical challenges in the diagnosis and treatment of atherosclerotic cardiovascular disease (ASCVD). Specifically, we will focus on the early detection of heart and kidney damage, the classification of disease risk types, and the understanding of underlying mechanisms driving disease progression. This research will also explore the development of AI tools that could significantly improve the accuracy of ASCVD diagnosis and treatment.
The need for this research arises from the limitations of current medical tests, which often assess heart and kidney health in isolation. Many ASCVD patients experience the interplay of multiple organ systems, resulting in a high rate of misdiagnosis. By combining heart MRI scans and kidney function data, along with genetic, lifestyle, and other factors, we aim to improve diagnostic accuracy and offer earlier interventions. Our research will also explore modifiable risk factors, such as specific metabolic abnormalities, which could provide new therapeutic targets to protect both heart and kidney function.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/decoding-causal-mechanisms-and-multifactorial-synergies-in-neuropsychiatric-and-chronic-disease-pathogenesis-and-multimorbidity-trajectories

Decoding Causal Mechanisms and Multifactorial Synergies in Neuropsychiatric and Chronic Disease Pathogenesis and Multimorbidity Trajectories

Last updated:
ID:
784989
Start date:
17 September 2025
Project status:
Current
Principal investigator:
Dr Zhe Chen
Lead institution:
Guangzhou Medical University, China

Current clinical management of multimorbidity remains reactive and symptom-focused due to limited understanding of cross-disease mechanisms. The UK Biobank’s multi-omics data (genomics, proteomics, imaging) and longitudinal phenotyping uniquely enable us to distinguish genetic predisposition from environmental mediation through instrumental variable analysis and resolve cytokine/neurotransmitter crosstalk between neuropsychiatric-chronic disease pairs via multi-layer network modeling. The paradigm shift from single-disease to pathway-centric frameworks could reveal novel prevention targets reducing comorbidity incidence, enable risk-stratified treatment matching using predictive algorithms and provide biological evidence for environmental policy interventions.
Our study aims to unveil potential causal factors and genetic biomarkers associated with neuropsychiatric disorders and chronic diseases, elucidate the underlying molecular mechanisms, thus propose novel preventive strategies. Additionally, our team aims to explore the underlying genetic landscape and biological correlations in order to uncover shared biological pathways and therapeutic targets for multimorbidity patterns and subsequently guiding clinical decision-making. Also, data models will be developed utilizing genetic biomarkers previously discovered and clinical features to facilitate disease diagnosis and predict treatment responsiveness in patients. This endeavor holds promise for enhancing disease management, minimizing notable side effects, and reducing healthcare costs linked to ineffective treatment protocols.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/decoding-multimorbidity-a-pathway-to-personalised-prevention

Decoding Multimorbidity: A Pathway to Personalised Prevention.

Last updated:
ID:
435869
Start date:
4 February 2025
Project status:
Current
Principal investigator:
Dr Artitaya Lophatananon
Lead institution:
University of Manchester, Great Britain

1. Explore the role of established risk factors and pathways for multimorbidity (e.g., cardiovascular disease, diabetes, hypertension, chronic respiratory diseases, cancer, osteoarthritis, chronic kidney disease, mental health disorders, neurological disorders, and dementia) in the UK population.
2. Discover the non-modifiable risks, lifestyle-related risk factors, medical history-related risk factors, imaging-related risks, genetic predispositions and metabolomic profiles, and explore the causal risks and pathways in multimorbidity (e.g., cardiovascular disease, diabetes, hypertension, chronic respiratory diseases, cancer, osteoarthritis, chronic kidney disease, mental health disorders, neurological disorders, and dementia) in the UK population.
3. Establish a comprehensive network analysis in the UK Biobank cohort by integrating current risk factors, genomic biomarkers and metabolomic profiles-related risks for multimorbidity (e.g., cardiovascular disease, diabetes, hypertension, chronic respiratory diseases, cancer, osteoarthritis, chronic kidney disease, mental health disorders, neurological disorders, and dementia).


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/decoding-residual-cardiovascular-risks-across-primary-and-secondary-prevention-a-multidimensional-analysis-of-genetic-proteomic-environmental-clinical-and-lifestyle-determinants

Decoding Residual Cardiovascular Risks Across Primary and Secondary Prevention: A Multidimensional Analysis of Genetic, Proteomic, Environmental, Clinical and Lifestyle Determinants.

Last updated:
ID:
786300
Start date:
4 August 2025
Project status:
Current
Principal investigator:
Dr Wenbin Zhang
Lead institution:
Zhejiang University, China

Questions
Despite advancements in prevention and treatment, cardiovascular diseases (CVD) remain the leading cause of mortality worldwide. Residual cardiovascular risk persists in secondary prevention, where optimal management of traditional factors like low-density lipoprotein cholesterol still leaves patients vulnerable. This residual risk is driven by genetic predispositions, inflammation, and lifestyle factors. Understanding these is equally crucial in primary prevention, where early identification and intervention can reduce long-term cardiovascular burden.

Objectives
1. Identify residual cardiovascular risk factors beyond traditional metrics, including genetic, environmental, lifestyle, and proteomic biomarkers.
2. Assess the predictive value of advanced omics data (e.g., proteomics) and imaging phenotypes for cardiovascular risk stratification.
3. Examine the role of emerging risk factors in primary and secondary prevention to guide early prevention and clinical application.
4. Develop integrative risk models combining genetic, proteomic, environmental, and lifestyle data for personalized prevention strategies.

Scientific Rationale
The UK Biobank offers a unique platform for investigating cardiovascular risk, with longitudinal data on genomics, proteomics, imaging phenotypes, and lifestyle metrics. These resources enable integrative analyses to uncover novel risk factors, elucidate mechanisms, and improve prediction models.

Proteomics identifies biomarkers linked to inflammation, fibrosis, and metabolic dysregulation, while imaging captures cardiovascular structural and functional changes. Together with genetic predispositions and lifestyle factors, these data comprehensively explore residual cardiovascular risk in primary and secondary prevention. Leveraging this multidimensional dataset, the study aims to refine prevention strategies, identify at-risk individuals earlier, and improve long-term cardiovascular outcomes.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/decoding-shared-mechanisms-multi-dimensional-genetic-and-multi-omic-drivers-of-chronic-disease-comorbidity-relationships

Decoding Shared Mechanisms: Multi-Dimensional Genetic and Multi-Omic Drivers of Chronic Disease Comorbidity Relationships

Last updated:
ID:
960873
Start date:
6 August 2025
Project status:
Current
Principal investigator:
Dr Yifeng Ren
Lead institution:
Chengdu University of Traditional Chinese Medicine., China

Research Question: How do genetic variation, multi-omics profiles (including transcriptomics, proteomics, metabolomics, microbiome), and environmental/lifestyle exposure risk factors interact to drive the development and co-occurrence (comorbidity) of major chronic diseases (specifically lung diseases, neurodegenerative/psychiatric disorders, cardiovascular diseases, diabetes, kidney diseases, cancers) and influence aging trajectories and mortality?
Objectives: Identify shared molecular pathways underlying chronic disease comorbidities. Investigate the chronic disease Comorbidity axis, including treatment effects and genetic targets. Develop integrated comorbidity prediction models using genetics, exposures, and multi-omics. Assess generalizability of findings across diverse ancestries in UK Biobank.
Scientific Rationale: Chronic diseases cause a global health burden and frequently co-occur, suggesting shared etiologies. While genetics contribute, interactions between genetic variation, environmental exposures, and dynamic molecular processes (reflected in multi-omics) in driving comorbidity remain poorly characterized. Notably, individuals with identical genetic mutations show variable disease severity/progression, highlighting the role of modifiable factors and molecular context. The UK Biobank provides an unprecedented resource to address this complexity through deep phenotyping, genetic data, and emerging multi-omics for ~500,000 individuals. This enables moving beyond single-disease studies to map the interconnected “diseaseome”. A critical gap exists in understanding the lung-brain axis. Observational associations exist, but causal mechanisms, shared pathways, and impacts of lung treatments on neurodegeneration are unclear. Similarly, shared etiology across other chronic diseases requires systematic multi-omics exploration across diverse populations to ensure equitable insights. This project harnesses UK Biobank’s scale and depth to fill these gaps.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/decoding-the-non-coding-genome-using-deep-neural-networks-and-high-resolution-functional-genomics

Decoding the non-coding genome using deep neural networks and high resolution functional genomics

Last updated:
ID:
295265
Start date:
27 May 2025
Project status:
Current
Principal investigator:
Professor James OLIVER JOSHUA Davies
Lead institution:
University of Oxford, Great Britain

Only 5% of the human genome encodes proteins, which are the molecules that are responsible for the way in which cells, tissues and organism’s function. The remainder of the genome determines when and how much of each protein should be made in different cell types. We have been able to interpret the protein coding part of the genome since the 1960s because the same code is used in all living organisms. However the non-coding genome is much more difficult to interpret because every cell type in every organism reads it as a different language. Our project aims to develop artificial intelligence based models to interpret the 95% of the genome that creates the instructions for complex life.
We plan to use these models to interpret the genome sequencing datasets in UK biobank to find new genes that are associated with human phenotypes and disease. In addition, the large datasets in the UK biobank could be used to test whether our models are accurate.
The project has the potential to impact at several different levels. First it might allow improved correlation between genotype and disease, allowing us to improve our ability to identify people at a high risk of developing disease. It is also likely to identify new genes and biological pathways that are important in developing disease and modulating disease outcomes, which will potentially have an important impact on drug discovery.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/decoding-the-pathogenesis-of-digestive-disorders-and-developing-predictive-tools-using-phenotypic-and-genomic-data-from-the-uk-biobank

Decoding the pathogenesis of digestive disorders and developing predictive tools using phenotypic and genomic data from the UK Biobank

Last updated:
ID:
157997
Start date:
15 February 2024
Project status:
Current
Principal investigator:
Dr Qian Cao
Lead institution:
Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, China

Many digestive disorders, such as inflammatory bowel disease and non-alcoholic fatty liver disease, are complex disorders with remarkable heterogeneity. Emerging evidence indicated that environmental and genetic factors are major contributors and exert their effects synergistically on the disease onset and progression. Currently, the underlying mechanisms remain elusive, and consequently, such diseases still place a huge economic burden on modern society. However, with the advent of the population-level large-scale prospective cohorts and the development of high-throughput technologies, we are now equipped with multiple powerful tools and are able to disentangle this by integrative analysis of both phenotypic and genomic/transcriptomic data. In this project, our interdisciplinary team will use clinical epidemiology approaches as well as molecular epidemiology methods to examine the following key questions in the GI field: (1) to identify the modifiable risk factors and biomarkers to elucidate their (moderating) functions on disease development; (2) to develop a set of predictive tools for the prediction of disease onset, disease progression, and etc., for the early detection of high-risk population; (3) to discover new genetic variants and candidate genes associated with digestive disorders and phenotypic traits; (4) to uncover the genetic correlations among digestive disorders and other complex diseases and their comorbidities; (5) to infer potential causal associations between modifiable risk factors and the digestive disorders; (6) to provide mechanistic insight into how the candidate genes in specific cell types exert their effects on disease development; (7) to identify potential molecular targets for drug discovery and drug repurposing. Our combinatorial and systematic approaches will allow us to develop user-friendly prognostic algorithms and interfaces, generate summary-level data for the GI community, and have a better understanding of disease pathogenesis and drug treatment for the benefits of both primary prevention in general practice and secondary/tertiary prevention for hospitalized patients. The findings from this study will aid health-care providers to identify high-risk individuals for certain digestive disorders, and ultimately develop personalized treatment accounting for genetic backgrounds.This research project is computational-demanding, we anticipate 36 months but depending on the outcomes, we may request an extension.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/decomposition-of-phenotype-dependent-pleiotropic-effects

Decomposition of phenotype-dependent pleiotropic effects

Last updated:
ID:
54148
Start date:
17 February 2020
Project status:
Current
Principal investigator:
Professor Xionglei He
Lead institution:
Sun Yat-Sen University, China

The aim of this project is to know how genetic pleitropy could shape the evolution of human population. Pleiotropy refers to the phenomenon that a single gene can affect multiple seemingly unrelated traits. It is known that pleiotropy is common. For example, in humans the deficiency of phenylalanine hydroxylase causes mental retardation, eczema, and pigment defects; a quantitative trait analysis in mice found that each quantitative trait loci affected ~ on average eight skeletal characters. Because of pleiotropy, the effect of genetic loci on a given trait may be contingent on other phenotypes. For example, smoking or being overweight increases cancer risk of cancer driver mutations; baldness appears more often in male than in female. Therefore, it’s difficult to understand pleiotropy by considering individual traits separately. Using the multi-dimensional omics data from UK Biobank, we can track the dynamics of allele frequency in multiple phenotypic backgrounds, by constructing and integrating the genotype-phenotype and phenotype-phenotype interaction network. We plan to finish the project within two years after we obtain the data. This study would provide novel insights into the co-occurrence or mutual-exclusiveness of diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deconstructing-the-genetic-basis-of-human-complex-phenotypes

Deconstructing the genetic basis of human complex phenotypes

Last updated:
ID:
474802
Start date:
6 December 2024
Project status:
Current
Principal investigator:
Mr Hakhamanesh Mostafavi
Lead institution:
NYU Grossman School of Medicine, United States of America

Most human traits and disorders, such as body weight and cardiovascular disease, are complex or polygenic, influenced by numerous genetic and environmental factors. Although Genome-Wide Association Studies (GWAS) have identified thousands of genetic variants associated with a variety of phenotypes, the functional mechanisms of these variants are still poorly understood, and extracting biological insights from these findings remains a challenge.

Our research aims to elucidate the genetic basis and biological mechanisms of complex traits and diseases by developing novel conceptual models and statistical methods to analyze genotype-phenotype data. We will use the UK Biobank resource to address four key areas:

1. Identification of Key Disease Genes: Most GWAS variants are non-coding with unknown target genes, and the relevance of many implicated genes remains unclear. We aim to develop methods to identify key disease-relevant genes, improving our understanding of biological processes and guiding therapeutic target discovery.

2. Mechanistic Pathways from Genotype to Phenotype: We seek to understand how genetic perturbations propagate through molecular and physiological pathways to affect organismal phenotypes and how phenotypic heterogeneity maps to genetic architecture.

3. Gene-Environment Interactions: We will explore how genetic effects are modulated by environmental factors and identify the genes and pathways involved in these interactions.

4. Improving Polygenic Risk Prediction: We aim to improve the accuracy, biological interpretability, and equitable use of polygenic risk scores for disease prediction.

To accomplish these objectives, we will conduct broad analyses across multiple traits and disorders to derive generalizable principles, as exemplified in our recent study (Mostafavi et al., 2023, Nat. Genet.), and focused analyses on specific phenotypes to uncover biological insights applicable to other traits (e.g., Mostafavi et al., 2020, eLife).


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deconvolution-of-factors-that-interfere-with-accurate-biomarker-interpretation

Deconvolution of factors that interfere with accurate biomarker interpretation

Last updated:
ID:
98959
Start date:
25 January 2023
Project status:
Current
Principal investigator:
Mr Jeff Cole
Lead institution:
Alden Scientific Incorporated, United States of America

Precision medicine is based on predicting, preventing and treating diseases in an individualized manner. For example, patients may have similar symptoms but need very different treatment. Some diseases are hard to follow up with treatment responses with clinical exams alone. To help physicians in these cases, a lot of effort has been spent on identification of blood markers that could predict disease diagnosis, disease progression and response to treatment. If successful, these blood markers make treating patients much easier and effective. However, establishing these blood markers for many diseases have been challenging. One of the main issues is identifying blood markers that replicate across multiple studies and centers. Our group hypothesizes that one reason for that is that “normal life” affects the blood levels of these biomarkers. For example: behaviors (nutrition choices, supplements, exercise routine, sleep hygiene); biological cycles (menstruation, circadian rhythm); and acute variances on day of draw (time of the day, last meal, time since wake), among others. There is an emerging body of scientific evidence supporting our hypothesis: (1) Multivitamin targeting hair, skin and nails have high concentrations of biotin, which is known to directly interfere with tests for markers of cardiovascular risk, hormone tests, and tests detecting infectious diseases; (2) Glucose levels are affected by circadian clock (the internal process that regulates the sleep-wake cycle of our bodies); (3) Menstrual cycle may affect cortisol levels, the classic stress response hormone; (4) Sedentary individuals with normal glucose and triglycerides may still be at risk for metabolic diseases, as indicated by an increase in triglyceride levels post-meal.

To investigate whether daily variances may indeed affect biomarkers levels, we will explore the correlation between individuals’ characteristics and habits with blood markers. Once we understand which factors affect these blood markers the most, we will develop methods to correct for these variances. We predict these corrections will lead to more accurate and stable markers.

The project should last about 2 years:
1- The first draft of correlations: 3 months
2- Deeper understanding of pre-selected correlations : 3 months
3- Algorithm to account for top 10 correlations: 6 months
4- Investigate accuracy of corrections on new data: 6 months
5- Reiterate steps 2/3/4 as needed : 6 months


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-and-frequent-phenotyping-study-2018

Deep and Frequent Phenotyping Study (2018)

Last updated:
ID:
40876
Start date:
12 February 2020
Project status:
Closed
Principal investigator:
Dr Vanessa Raymont
Lead institution:
University of Oxford, Great Britain

Alzheimer’s disease remains one of the most common unmet medical needs today. Without a single treatment for the disease process itself, clinical trials are failing almost certainly because they are performed too late in the disease process. Trials need to be performed in the preclinical or prodromal phase but such designs necessitate the use of biomarkers. The purpose of our study is to provide such biomarkers. Success in doing so will speed trials, reduce costs, increase productivity and ultimately contribute to more effective pipelines for early clinical drug development. This aligns closely with UK Biobank’s goal of improving prevention and treatment of dementia.

We plan to enlist 250 people into the study, 200 with AD before symptoms have started. We seek to work with UK Biobank to identify these people in a screening process making use of data already collected in UKBiobank, with the permission of participants. Those people meeting the study inclusion criteria will have a series of tests including PET and MRI imaging, other brain scans (EEG and MEG), measures in the eye, of gait and movement, and of cognition using standard approaches and twith connected devices including smart phones. In addition participants will have blood tests and spinal fluid tests and all of these will be repeated many times over the period of one year. These data will be used to find a biomarker of progression to be used in clinical trials of potential therapies.
Given the intensive nature of the proposal, we conducted a six month pilot study in people with mild memory problems. All assessments were completed with the exception of repeat lumbar puncture where headache was encountered at baseline (an expected occurrence in less than 10% of people), in one participant who took an unanticipated holiday, and in a small number of cases of equipment failure. We worked closely with the Alzheimer’s Society who formed a stakeholder consultation group and conducted participant feedback studies using questionnaires, focus groups and individual interviews. The feedback received was universally positive, no participants found any unacceptable and the stakeholder consultation group helped to inform protocol development.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-algorithms-based-on-ocular-and-health-status-predict-cognitive-dysfunction-and-development-of-dementia

Deep learning algorithms based on ocular and health status predict cognitive dysfunction and development of dementia.

Last updated:
ID:
75750
Start date:
9 June 2023
Project status:
Current
Principal investigator:
Dr Katerina Kiroshka
Lead institution:
The Fund for Medical Research Infrastructure By Barzilai Medical Center, Israel

Worldwide, around 50 million people have dementia. The prevalence of dementia is estimated at 10% of individuals aged 65 and older. Its incidence increases with age, from 5% in ages 65-74 to 30% in ages 85 and older. (WHO)
We review two major risk factors for developing dementia. First, visual impairment that affects productivity and significant lifestyle changes in elderly population. Moderate-to-severe visual impairment is a potential predictor and a risk factor for dementia. We study the most prevalent reason for visual impairment among the aging population, and the potential interaction between visual impairment and dementia.
Second, cardiovascular risk factors are also suggested risk factors for dementia. Multiple studies conclusively show that the significant risk factors for stroke are also the most common risk factors for cardiovascular and peripheral vascular disease, suggesting that these disorders share a common mechanism of vascular injury. Risk factors for cardiovascular and peripheral vascular disease due to SCORE quiz (age, sex, systolic blood pressure, use of antihypertensives, left ventricular hypertrophy, prevalent cardiovascular disease, smoking status, atrial fibrillation, and diabetes mellitus) Vascular factors commonly lead to a spectrum of asymptomatic brain injury and potential dementia.
Our aim is prediction and early detection of dementia which are very important to early and optimal management. We estimate algorithms based on deep learning that can be applied to extract novel information such as dementia from ocular and health status.
Deep learning is a class of machine learning techniques that has tremendous global interest in the last few years, recognizing that it is increasingly embraced and utilized.
In medicine and healthcare, deep learning (DL) has been primarily applied to medical imaging analysis, in which DL systems have shown robust diagnostic performance in detecting various medical conditions. Major ophthalmic diseases which DL techniques have been used for including diabetic retinopathy (DR), glaucoma, age-related macular degeneration (AMD). DL has also been applied to estimate refractive error and cardiovascular risk factors (eg, age, blood pressure, smoking status and body mass index).


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-and-genome-wide-analysis-to-determine-the-role-of-reticular-pseudodrusen-in-the-progression-of-age-related-macular-degeneration

Deep learning and genome-wide analysis to determine the role of reticular pseudodrusen in the progression of age-related macular degeneration

Last updated:
ID:
60078
Start date:
27 July 2020
Project status:
Current
Principal investigator:
Mr Adnan Tufail
Lead institution:
Moorfields Eye Hospital NHS Foundation Trust, Great Britain

Brief background and need for the research:

Age-related macular degeneration (AMD) is a leading cause of blindness and is characterised in its early stage by deposits under the macula called drusen. Over time, many eyes develop atrophy (worn out patches of the retinal layer that picks up light), also called “dry AMD”, for which there is no treatment. A minority of eyes with AMD develop a secondary complication of abnormal blood vessels growth at the back of the eye (“wet AMD”) which leads to leakage of fluid into or under the retina. There is currently treatment for wet AMD. It is imperative to better understand the biological pathways and genetic associations that are linked to dry AMD to provide a suitable treatment in the future.

Historically, drusen are hallmarks of AMD. However, reticular pseudodrusen (RPD) are another form of abnormal tissue deposits that can mostly be seen in patients with AMD but also with some other conditions that involve the back of the eye. Recently, RPD have been recognised as an important risk factor for advanced AMD, and possibly a greater risk factor than the presence of conventional drusen deposits alone. There is limited understanding of how RPD drives disease.

The UK Biobank is an existing large database of patient images and genetic data that have got individual patient consent for research use.

Aim of the study:
The proposed study addresses important gaps in our knowledge of the key factors for advanced AMD by first developing image recognition software (artificial intelligence tools) to automatically detect RPD. We then intend to apply the software developed in the previous step to detect RPD in existing patient images from the Biobank. The last step will be diagnosing genetic data from the Biobank, to see if previously undiscovered genetic risk factors can be detected.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-and-genomic-analyses-of-functional-genetic-variants-and-region-specific-genes-in-human-diseases

Deep learning and genomic analyses of functional genetic variants and region specific genes in human diseases

Last updated:
ID:
256056
Start date:
11 November 2024
Project status:
Current
Principal investigator:
Professor Ming Zhang
Lead institution:
Tongji University, China

Understanding the human genome is pivotal for personalized medicine and improving disease treatment. But the functional roles of noncoding variants and structural variants in our DNA are still largely unclear. In addition, the link of spatially specific genes and diseases is largely unknown. Understanding these functional variants and region-specific genes could hold the key for smart diagnosing and treating diseases.

Deep learning is a type of artificial intelligence that mimics the way humans learn. Some deep learning models have been designed to predict the functional roles of genetic variants, such as pre-mRNA splicing and gene expression. Despite their potential, current models often rely on a narrow selection of genetic data for training, severely undercutting the rich diversity within human genetics.

Our current research proposal leverages the UK Biobank’s genetic and health data, including whole-genome sequencing (WGS) data from around 500,000 individuals. This unique resource offers us an opportunity to develop novel deep learning models that can better predict the functional roles of genetic variants, especially those in the noncoding and structural regions of the genome. Moreover, the resource would allow us to identify the link between region specific genes and disease risk.

In a duration of 3 years, our research aims to develop several deep learning models that can predict how genetic variations affect RNA splicing, how genetic variants affect RNA alternative polyadenylation sites, and how genetic variants affect DNA methylation status. In addition ,our proposal would pinpoint the roles of region specific genes in the etiology of human diseases.

By predicting these functional effects, we hope to unravel how noncoding and structural variants affect key biological processes and establish their links to complex human diseases (such as neurodegenerative disease, cardiovascular diseases and cancer). Our methods involve using the UK Biobank’s genetic sequences to train these models, ensuring they can handle the diversity and complexity of human genetics more effectively.

In navigating the intricate roles of our genetic code, we could unlock precise treatments and diagnostics. In addition, we aim to provide novel insights into genetic mechanisms of noncoding variants and structural variants underlying human diseases, as well as the roles of region specific genes underlying human diseases. By understanding the links of functional/regional genetic elements and biomarkers, we could significantly enhance our ability to predict, prevent, and treat various diseases, tailoring our approaches to personalized medicine.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-assisted-diagnosis-of-obstructive-sleep-apnea-osa-based-on-multimodal-data

Deep Learning-Assisted Diagnosis of Obstructive Sleep Apnea (OSA) Based on Multimodal Data

Last updated:
ID:
575800
Start date:
1 June 2025
Project status:
Current
Principal investigator:
Dr Jun Tai
Lead institution:
Capital Institute of Pediatrics, China

1. Background
Obstructive Sleep Apnea (OSA) is a common sleep-related breathing disorder characterized by recurrent partial or complete obstruction of the upper airway during sleep, leading to reduced oxygen saturation, poor sleep quality, and daytime sleepiness. Traditional OSA diagnosis mainly relies on polysomnography (PSG) and clinical evaluations, but these methods can be time-consuming, subjective, and less accurate for diagnosing mild or moderate cases of OSA.
With the development of deep learning techniques, multimodal data-based methods present new opportunities for improving OSA diagnosis. By combining various physiological signals (e.g., airflow, blood oxygen saturation, electroencephalography) with imaging data (e.g., CT or MRI scans of the upper airway), deep learning models can significantly enhance diagnostic accuracy and efficiency.
2. Objectives
This study aims to develop a deep learning-based system for the auxiliary diagnosis of OSA, using multimodal data sources from the UK Biobank, including sleep monitoring data, imaging data, clinical data, and genetic data. The goal is to improve early OSA diagnosis and provide.
3.Scientific Rationale:
OSA is a complex condition influenced by various factors, including anatomical, physiological, and genetic components. Current diagnostic methods, such as polysomnography (PSG), are often costly and inefficient, particularly for diagnosing mild or moderate cases. Integrating multiple data sources through deep learning can address these limitations. By using data from the UK Biobank, which includes sleep monitoring, imaging, clinical, and genetic information, this project aims to enhance diagnostic accuracy and provide insights into the genetic predispositions of OSA.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-autoencoders-for-learning-lower-dimensional-manifolds-of-structural-and-functional-neuroimaging-data

Deep Learning Autoencoders for learning lower-dimensional manifolds of structural and functional neuroimaging data

Last updated:
ID:
30707
Start date:
10 August 2021
Project status:
Closed
Principal investigator:
Professor Tim Hahn
Lead institution:
Institute for Translational Psychiatry, Germany

When analyzing the brain through MRI scans, researchers are typically faced with up to a half a million data points. While this is impossible to analyze for humans, even algorithms from artificial intelligence can get overwhelmed by the mere amount of data. Therefore, insights from neuroimaging data can currently not be translated into useful information for the clinical practice, e.g. to predict the best treatment, the best drug or help with difficult diagnosis. Typically, this problems can be tackled in two ways: one is to scan millions of subject’s brains, and second, is to compress the information contained in the brain scans to a more useful representation. We intend to use a technique called Autoencoder, which represents a data-driven compression method. This would enhance the algorithm’s ability to deliver clinically relevant predictions while it would also substantially decrease the number of research subjects needed for such research in the future. Thus, the UK Biobank neuroimaging data provides the unique opportunity to solve one of the fundamental problems in contemporary machine-learning on neuroimaging data. The project is expected to take two and a half years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-based-analysis-of-digital-phenotypes-and-response-evaluation-for-bipolar-disorder

Deep Learning Based Analysis of Digital phenotypes and Response evaluation for Bipolar disorder

Last updated:
ID:
97674
Start date:
5 June 2023
Project status:
Closed
Principal investigator:
Mr Anas Turki Althobaiti
Lead institution:
Northumbria University, Great Britain

Identify digital markers capable of discriminating bipolar disorder and predicting response to long-term treatment. Develop a deep learning-based analysis approach for the detection of bipolar disorder. Explore novel, diverse biomarkers, investigate newly inducted biomarkers. Formulate algorithm for computing the effectiveness of individual biomarkers using different feature selection methods.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-based-analysis-of-retinal-oct-scans-for-detection-of-alzheimers-disease

Deep Learning-Based Analysis of Retinal OCT Scans for Detection of Alzheimer’s Disease

Last updated:
ID:
82266
Start date:
15 February 2022
Project status:
Current
Principal investigator:
Mrs Yasemin Turkan
Lead institution:
Isik University, Turkiye

Alzheimer’s disease (AD) is the most common form of dementia. It is an irreversible, progressive brain disorder marked by a decline in cognitive functioning with no treatment. It is characterized by a massive decrease in brain size due to the accumulation of proteins (amyloid-beta and tau ) in the neurons. Eyes extend the brain as both the retina and brain grow from the same neural tube. Postmortem studies in AD also highlighted the collection of these proteins in the retina. More recently, high-resolution visual imaging techniques, including optical coherence tomography (OCT), have been proposed as tools for evaluating structural changes in the retina of AD patients. It is
Conventional diagnostic methods from medical images greatly depend on physicians’ professional experience and knowledge. Artificial intelligence (AI) has improved the performance of many challenging tasks when working with high-resolution, complex imaging data. Artificial neural networks are a subset of AI inspired by a simplification of neurons and their connections in the brain. Deep learning (DL) is a multi-layer structure of neural networks that mimics human learning by analyzing data with a given logical structure.
This project is for a Ph.D. thesis research planned for three years, focusing on using deep learning-based analysis of retinal OCT scans for AD detection. Even though this technique is widely used to detect many other retinal diseases from OCT images, there is no application in AD.
Retinal scans obtained from OCTS devices are two-dimensional and three-dimensional images. Despite their high performance, DL architectures are black-box models. Trusting their predictions is an important factor in using them for decision-making in medicine. Therefore, this research aims to train the model with retinal images and develop algorithms that will help clinicians to review and visualize the decision process. The power of explainability tools can also help to highlight relevant patterns and even discover new ones.
It is not easy to collect sufficient, high-quality, and uniformly annotated data to build high-accuracy models. Research studies exclude old patients with multiple retinal illnesses. Therefore, only a minimal subset of collected data could be used in the studies. We also plan to investigate various learning methods to increase learning with small AD datasets by transferring knowledge from other studies and datasets.
This research aims to create an efficient and explainable model that will learn to classify stages of Alzheimer’s disease from the currently used retinal scans.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-based-application-of-imaging-genomics-in-brain-diseases

Deep-learning-based application of imaging genomics in brain diseases

Last updated:
ID:
75310
Start date:
1 November 2021
Project status:
Current
Principal investigator:
Professor Shuqiang Wang
Lead institution:
Shenzhen Institutes of Advanced Technology, China

Aim:The project aims to develop a computer method for predicting brain disease from multi-modal brain image and genomic data. This method can explore the mechanisms of brain diseases and provide biomarkers for treatments.
Scientific rationale: Patients with different kinds of cognitive disorders differ in the brain structure, functional connections and brain tissue. For example, the volume changes in hippocampus brain region is proved to be closely correlated with the progression of Alzheimer Disease; the functional connections in patients with cognitive disease is very different compared with the normal people. All these changes can be observed by the neuroimaging techniques, such as fMRI ,DTI and T1-weighted MRI. In addition, the cognitive disease is greatly affected by genetic inheritance, and studies have confirmed that there are multiple gene loci strongly correlated with changes in hippocampus volume. Therefore, through the analysis of brain images and gene data, we can study the mechanism of brain circuits for the treatment of brain cognitive diseases, and provide biological markers for the diagnosis and early treatment of diseases.
Project duration:The project will last for 3 years
Public health impact:This project will produce a new computational method for diagnosis, typing, prediction and prevention of brain diseases. The results of this project will probably provide substantial new insights into the genetic landscape of the brain and offer a scientific value that could advance application on normal brain development and neurological disorders and improve the diagnosis of brain disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-based-causal-inference-methods-for-variant-interpretation

Deep learning based causal inference methods for variant interpretation

Last updated:
ID:
79957
Start date:
10 March 2022
Project status:
Current
Principal investigator:
Professor Wing Hung Wong
Lead institution:
Stanford University, United States of America

A generic question in genetics is whether a non-coding variant affects a particular disease. Genome-wide association study (GWAS) is an approach used in genetics research to associate specific genetic variations with particular diseases. However, correlation does not imply causation.Therefore, it is hard to interpret the GWAS results. Thanks for the recent breakthroughs in combination of deep learning and statistics, we try to examine the effects of the genetic variants that are associated with the biomarker on the disease outcomes. If the individuals with the genetic variants associated with specific biomarkers are observed to have increased disease risk, one could infer that the biomarker is causally linked to the disease. Here, genetic variants are ideal choices of instrumental variables because they are inherited from parents and are not subject to the influences of confounding variables.

Inferring causal relationships is a more significant theoretical and computational challenge than measuring correlations, so our model will combine casual inference theory and deep learning technique. We expect to produce more accurate, robust, and equitable tools for genetic prediction of disease risk. As a basic methodological tool, it will enable individual level disease risk estimation and early interventions for complex diseases such as cancer, hypertension, and cardiovascular disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-based-chromatin-accessibility-snp-targeting-for-increasing-gwas-power

Deep learning-based Chromatin Accessibility SNP Targeting for increasing GWAS power

Last updated:
ID:
786815
Start date:
4 September 2025
Project status:
Current
Principal investigator:
Professor Valentina Boeva
Lead institution:
ETH Zurich, Switzerland

Genome-wide association studies (GWAS) have transformed our understanding of complex traits but are often limited by a high multiple-testing burden and the inclusion of many non-functional, non-coding variants. Our method seeks to enhance GWAS power by pre-filtering SNPs based on their predicted impact on chromatin accessibility, a key indicator of regulatory function. Utilizing the deep learning capabilities of the Enformer model, we compute SNP Activity Difference (SAD) scores that measure the effect of individual genetic variants on chromatin openness, thereby prioritizing SNPs that are likely to influence gene expression. This approach addresses the critical research question of whether integrating functional annotations can improve the recovery of true genetic associations and reveal novel loci that might be missed by traditional GWAS methods. Our objective is to systematically reduce the number of independent tests by excluding SNPs with negligible regulatory impact, allowing for a less stringent significance threshold and, ultimately, increasing the power to detect meaningful associations. By linking genetic variation directly to regulatory mechanisms, our method not only streamlines the analytical pipeline but also enhances the biological interpretability of GWAS findings, paving the way for deeper insights into the genetic architecture of complex traits.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-based-genetic-analysis-and-phenotype-prediction-using-variants-of-any-frequency-based-on-uk-biobank-sequencing-datasets

Deep-learning-based genetic analysis and phenotype prediction using variants of any frequency based on UK Biobank sequencing datasets

Last updated:
ID:
81358
Start date:
2 June 2023
Project status:
Current
Principal investigator:
Professor Oliver Stegle
Lead institution:
German Cancer Research Center (DKFZ), Germany

A key challenge in medicine is to understand how genetic variations, arising from mutations and inheritance patterns, determine medically relevant traits – such as cholesterol level – and predisposition to diseases such as cancer within populations. While the contributions of common genetic variants to different human traits have been extensively studied and characterized, there is still a lack of understanding of the impact of rare genetic variants. However, recent studies have shown that rare genetic variants strongly contribute to a variety of traits.

Our aim is to develop and apply novel computational methods that allow for identifying and quantifying the contribution of rare genetic variants in humans. The ultimate goal of this work is to integrate the effect of rare variants into models that can predict human disease traits.

The scale and diversity of the UK Biobank dataset offers the possibility to develop and build the proposed trait/disease prediction models, using advanced techniques of machine learning and artificial intelligence. The resulting models will be made publicly available for download and use by the scientific community. Moreover, the output of these models will yield new insights into the specific genes that are involved in different traits and diseases, including predisposition to severe COVID-19.

Collectively, this project will contribute to an improved understanding of the genetic basis of human disease traits, which will ultimately enable improved diagnosis and new treatment strategies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-based-multi-modal-medical-image-registration-and-segmentation

deep learning based multi-modal medical image registration and segmentation

Last updated:
ID:
89291
Start date:
11 October 2022
Project status:
Current
Principal investigator:
Professor Qing Pan
Lead institution:
Zhejiang University of Technology, China

1. using the multimodality brain imaging, we aim to develop novel multimodality registration algorithms using deep learning, which extract features from images and optimize the parameters.
The developed method could be used to perform planning before a brain resection surgery,
and later used for image navigation during a brain surgery.

2. to better understand brain functions, using brain parcellation which labeling the areas of brain and multi-atlas methods, perform brain function analysis
3. compare brain imaging with cognitive functions
4. the proposed project timeline would be 2-3 years, and hopefull a research-level pre-surgical imaging planning algorithm and software are developed in this project

The impact to public health:

In an aging society, this study could help to better understand brain development and cognitive functions, which could help to alleviate aging problems, and hopefully solve brain degenerative diseases such as Alzheimers’ , Parkinson’s diease, Epilepsy or depression

It would also help to predict brain disease outcomes before surgery, as well as to better evaluate the patient’s progression over a long period.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-based-multi-omics-imaging-and-pathology-data-fusion-for-predicting-immune-therapy-response-in-urological-cancers

Deep Learning-Based Multi-Omics, Imaging, and Pathology Data Fusion for Predicting Immune Therapy Response in Urological Cancers

Last updated:
ID:
705801
Start date:
22 April 2025
Project status:
Current
Principal investigator:
Mr Jie Ming
Lead institution:
Beijing Friendship Hospital, Capital Medical University, China

Research Question
Immune therapy, especially immune checkpoint inhibitors like PD-1/PD-L1 and CTLA-4 inhibitors, has shown significant efficacy in treating urological cancers, including bladder cancer, prostate cancer, and renal cell carcinoma. However, clinical responses vary greatly among patients, and traditional biomarkers, imaging techniques, and pathology data, often used in isolation, are limited in their ability to accurately predict treatment outcomes.

Research Objective
This study aims to integrate clinical information, multi-omics data (genomics, transcriptomics, proteomics), imaging data (CT, MRI, PET), and pathology data (tissue samples, immunohistochemistry) to improve the prediction of immune therapy responses in urological cancers. By combining these heterogeneous data sources, we seek to develop a more accurate and comprehensive predictive model. Traditional models relying on single data types often fail to capture the complexity of immune responses. Multi-modal data fusion through deep learning techniques will enable the extraction of complex features across data types, improving prediction accuracy.

Scientific Rationale
Immune therapy, particularly checkpoint inhibitors like PD-1/PD-L1 and CTLA-4 inhibitors, has demonstrated promising results in urological cancers. However, patient responses remain highly variable. Most existing studies rely on single-modality data, such as imaging or pathology alone, which limits their ability to fully capture the complexity of immune therapy outcomes. There is a clear need for more integrated, multi-modal approaches to enhance prediction accuracy.

Using deep learning-based multi-modal data fusion, we can analyze these diverse data types simultaneously, uncovering complex patterns missed by single-modality approaches. This leverages advancements in data availability, imaging, and machine learning, aiming to improve patient stratification and enable personalized treatment plans.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-based-multi-omics-research-for-discovering-metabolic-syndrome-and-related-biomarkers

Deep learning based Multi-Omics research for discovering Metabolic Syndrome and related biomarkers.

Last updated:
ID:
825589
Start date:
17 July 2025
Project status:
Current
Principal investigator:
Mr Dongwook Jeong
Lead institution:
Sungkyunkwan University, Korea (South)

Metabolic syndrome is a cluster of conditions involving various metabolic disorders, significantly increasing the risk of hyperlipidemia, diabetes, hypertension, obesity and cardiovascular disease. The global prevalence of metabolic syndrome is on the rise, with over one-third of the U.S. population affected. Given its complex cause of onset, it is likely that metabolic syndrome interacts with a wide range of other diseases.
In this study, we apply a generative deep learning model, Variational Autoencoder(VAE), to analyze proteomics and metabolomics data from individuals with metabolic syndrome. Our goal is to explore potential associations between metabolic syndrome and other diseases through multi-omics integration.
We have identified candidate biomarkers associated with metabolic syndrome using external cohort data and are currently planning to validate these findings in the UK Biobank dataset.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-based-multiclass-retinal-fluid-segmentation-and-detection-in-optical-coherence-tomography-images-using-a-fully-convolutional-neural-network

Deep-Learning based Multiclass Retinal Fluid Segmentation and Detection in Optical Coherence Tomography Images using a Fully Convolutional Neural Network

Last updated:
ID:
43402
Start date:
9 January 2019
Project status:
Closed
Principal investigator:
Professor Mirza Faisal Beg
Lead institution:
Simon Fraser University, Canada

Age-related macular degeneration (AMD) is the third leading cause of blindness in the world and the first in industrialized countries. Patients with AMD can suffer from blurriness and dark spots in their central vision. Diabetic retinopathy (DR) affects approximately one third of the people with diabetes, and in advanced stages it also leads to blurry vision and floating spots.

In these and other eye diseases, a major problem is fluid build-up and swelling inside the retina created by leaky blood vessels. Early detection of these fluids is important for successful treatment and management of the diseases.

Optical coherence tomography (OCT) provides 3D cross-sectional images of the retina and visualization of the retinal fluids. However, the areas of fluid build-up can be small and difficult to see without careful examination of each image, and this can be a costly and time-consuming process for clinicians with sometimes several hundreds of cross-sections to examine in each eye.

We have developed a novel framework using deep neural network (DNN) to automatically detect multiple types of fluid regions in OCT images, and identify eyes with specific fluid types. Our framework has achieved the highest level of accuracy for these tasks and won the first place in the 2017 MICCAI RETOUCH competition.

With our promising results, we hope to improve our framework in the next few years by training the DNN with a much larger number of data, including from UK Biobank, and develop a clinical software for ophthalmologists for accurate detection and detailed information of the retinal fluids, such as the shape and volume of the fluid regions. This tool has potential for helping ophthalmologists with early detection of harmful fluid-build ups in the retina, and tracking the disease progress and treatment effectiveness with accurate, quantitative information.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-based-multimodal-integration-of-imaging-and-genetic-features-for-carotid-atherosclerosis-characterization

Deep Learning-Based Multimodal Integration of Imaging and Genetic Features for Carotid Atherosclerosis Characterization

Last updated:
ID:
784342
Start date:
3 July 2025
Project status:
Current
Principal investigator:
Dr Yuhan Yang
Lead institution:
Beijing University of Chinese Medicine, China

Research Questions:What distinct imaging-genetic signatures differentiate high-risk carotid atherosclerosis phenotypes associated with neurological/cardiovascular events? Can AI-driven multimodal integration of ultrasound morphology, genomic data, and clinical variables improve risk stratification beyond conventional paradigms? How do genotype-phenotype interactions underlying plaque vulnerability inform novel therapeutic targets? This study addresses these gaps by synergizing deep learning-based image analysis, GWAS, and network medicine to enable precision risk prediction and intervention.
Objectives:This study aims to: (1) identify high-risk imaging-genetic signatures of carotid atherosclerosis through multimodal integration of ultrasound-derived features and genomic data; (2) develop an artificial intelligence (AI)-driven risk stratification model by synthesizing demographic, clinical, imaging, and multi-omics data; and (3) explore novel therapeutic targets by deciphering the phenotype-genotype interplay underlying carotid plaque vulnerability.
Scientific Rationale: Ultrasonographic metrics, including carotid intima-media thickness (cIMT) and plaque burden, have demonstrated incremental prognostic value for ischemic events beyond traditional risk factors. Concurrently, recent advances in biomedical big data analytics-encompassing high-resolution imaging, genomic profiling, and computational biology-enable systematic interrogation of atherosclerosis pathophysiology. Specifically, AI frameworks exhibit unique potential to decode complex associations between plaque morphology, genetic variations, and clinical outcomes through multimodal data fusion, thereby addressing critical gaps in current risk prediction methodologies. The findings from this project will be disseminated through peer-reviewed scientific publications in open-access journals in accordance with UK Biobank’s policies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-based-on-data-integration-identifies-potential-therapeutic-targets-in-neurodegenerative-disease

Deep learning based on data integration identifies potential therapeutic targets in neurodegenerative disease

Last updated:
ID:
83935
Start date:
12 December 2023
Project status:
Current
Principal investigator:
Professor S. Stephen Yi
Lead institution:
University of Texas (UT Austin), United States of America

For hard-to-treat neurodegenerative diseases, accurate classification and risk prediction will no doubt benefit diagnosis and improve patient life quality. Despite considerable efforts made, it remains difficult to effectively integrate different types of patient data available. To begin to solve this challenge, we plan to develop a deep learning-based, risk-associated prediction framework on neurodegenerative disease patients using coupled genomics and health-related data that include mutation profiles and MR imaging. We expect that our framework will achieve promising predictive performance in distinguishing high-risk versus low-risk patients, even with incomplete data available. Our study here will have major clinical implications as it may yield novel biomarkers or drug targets for a new generation of therapeutics in neurodegenerative disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-based-prediction-of-alzheimers-disease-using-genomics-and-neuroimaging

Deep-learning based prediction of Alzheimer’s disease using genomics and neuroimaging

Last updated:
ID:
56236
Start date:
4 August 2020
Project status:
Current
Principal investigator:
Professor Mirza Faisal Beg
Lead institution:
Simon Fraser University, Canada

Alzheimer’s disease (AD) is a neurodegenerative disorder, characterized by progressive cognitive decline and is the most common form of dementia. AD is an important reason for concern since it is affecting more and more people over time and no treatment is available yet. Brain cells are damaged during the process of developing AD and since this damage is irreversible, a huge focus is on predicting the onset of the disease. The goal is to find a reliable way to identify individuals who will probably develop AD as well as the time to conversion for those individuals and after achieving this goal, it will become possible to find and apply a treatment to prevent or slow down the brain tissue damage.

Alzheimer’s disease is a complex disease and depends on many factors. Genetic variants contribute to the risk of developing AD. Each of these genetic risk factors contributes to the disease risk in a different way. In some cases, the contribution to the disease risk will be small, while in other cases having a specific variant can have a large impact on the disease risk.

In this project we will design a deep neural network (DNN) to predict an individuals’ time to conversion to AD using combination genetic data, structural measurements extracted from image modalities such as MRI and clinical information gathered from different sources including UK Biobank. Our hypothesis for this project is that genetic markers will increase the accuracy of identifying those that convert to DAT. We hope to develop a clinical software in the next following years to predict the accurate time to conversion to AD that can facilitate the process of searching and applying the right cure at the right time.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-based-systemic-disease-screening-at-the-population-level-using-retinal-images

Deep Learning based systemic disease screening at the population level using retinal images

Last updated:
ID:
101900
Start date:
4 May 2023
Project status:
Current
Principal investigator:
Ms Pooja Barak
Lead institution:
LifeBytes India Private Limited, India

The eye is an important organ of the body that manifests changes in response to several systemic diseases. Several studies have attempted to characterize the retinal changes that occur in response to specific systemic disorders so that these changes may be used as markers for systemic diseases. Visualization of retinal alterations can be done by obtaining retinal photographs of patients that preclude the need for complex and/or invasive diagnostic procedures.

Systemic diseases are those that affect the entire body rather than just one organ or tissue. Such disorders include diabetes mellitus, hypertension, chronic kidney disease, rheumatoid arthritis, atherosclerosis, and metabolic syndrome. Most systemic diseases such as diabetes, arthritis, hypertension, and kidney disease are chronic and result in severe health consequences for the patients. Early detection of such conditions is important for optimal management through dietary and lifestyle changes resulting in better health outcomes for the patient.

The aim of this project is to characterize the retinal alterations associated with systemic diseases so that they may be used as early predictors of the disease. As the burden of systemic diseases is increasing globally, we need effective, accurate and cost-effective approaches to determine the susceptibility of patients to certain conditions. We also intend to characterize the underlying genetic changes that are associated with retinal changes and systemic diseases so that we can investigate important associations between retinal and genetic changes of a specific systemic condition.

Furthermore, this project will aim to develop a novel deep neural network (DNN) model to predict the risk of systemic diseases using both retinal images and genomic data. This model will consist of two parts: a classification network to identify high-risk patients and a regression network to assign a risk score. This will allow for more accurate and individualized predictions, taking into account both retinal and genetic factors.

Additionally, the project will include a population-based approach, including individuals from different ethnic backgrounds, to account for variations in retinal pigmentation and genetic makeup. This will ensure that the developed models are generalizable and applicable to a diverse population.

The ultimate goal of this project is to develop a non-invasive and cost-effective diagnostic tool for early detection of systemic diseases using retinal images and genomic data. The results of this study have the potential to greatly improve the management and outcome of chronic conditions and improve the quality of life for patients.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-for-causal-inference-on-genetic-and-medical-imaging-data

Deep Learning for Causal Inference on Genetic and Medical Imaging Data

Last updated:
ID:
568177
Start date:
11 October 2025
Project status:
Current
Principal investigator:
Mr Julius Herzig
Lead institution:
University of Hamburg, Germany

Observational data and the UK Biobank have large sample sizes but it is difficult to establish causality. Treatment assignment is unbalanced and confounding factors influence the outcomes. Advances in causal inference allow to control for confounding factors using machine learning methods for tabular data (doi:10.1111/ectj.12097). With the current work of our research group, DoubleMLDeep (doi:10.48550/arXiv.2402.01785) multimodal data can be directly used as a confounder in causal analyses. No manual feature engineering or expert knowledge on the multimodal data is required for analysis. This is promising as imaging provides detail on a person’s physical state that is not available in medical records traditionally used. Genetic data also contains information that is a confounder in many observational studies (e.g. risk of death depends on genes).
Following the development of methods that allow controlling for confounding using multimodal data, can they be used with medical data to improve the causal validity of findings? For this it needs to be verified if the networks are able to train well with the required modifications for causal inference and that the data used has sufficient predictive power.
Two potential applications are targeted for this evaluation:
Use of genetic sequencing data directly as a confounder to evaluate the impact of high blood pressure on cardiac outcomes (extension of doi:10.1001/jamacardio.2018.1717). Can feature engineering to identify important genes be replaced by passing the genetic information directly into a neural network?
Use of chest imaging data as a confounder in an observational assessment of the coronavirus vaccine effectiveness of preventing severe illness or death. The image contains a multitude of information about lungs and could be a good predictor for the outcome. Can confounding in a treatment assessment be better controlled using images rather than traditional use of preexisting conditions?


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-for-early-detection-of-the-structural-complications-of-myopia-that-can-lead-to-visual-loss-in-later-life

Deep learning for early detection of the structural complications of myopia that can lead to visual loss in later life.

Last updated:
ID:
273156
Start date:
29 October 2024
Project status:
Current
Principal investigator:
Mr Clement Kangombe
Lead institution:
Technological University Dublin, Ireland

Short sightedness (myopia) is one of the commonest eye conditions and by 2050, nearly 5 billion people will be affected. Most people think of myopia as a condition which needs correction with glasses or contact lenses. But in later life, myopia can lead to a range of complications which can leave the worst affected with significant loss of vision. In the over 75-year-old bracket, complications of myopia is the fourth commonest cause of vision impairment.
The main aim of this research is to develop a computerized system that provides eye care professionals with an early warning system of eyes that are at most risk of developing vision impairment due to myopia.
The duration of this project will be 3 years and it will be used towards my qualification for a PhD. The expected impact of this project will be to add new knowledge to the domain on detecting possible early signs of myopia using retinal images to help clinicians with the management of the disease.
In terms of public health impact, this project is designed to provide early detection of eyes that are of risk of vision loss due to the complications and help us to develop new treatments to keep short sighted eyes healthier as we age and prevent vision impairment in later life.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-for-predictive-modelling-of-alzheimers-disease-related-dementia

Deep learning for predictive modelling of Alzheimer?s disease related dementia

Last updated:
ID:
32575
Start date:
22 November 2018
Project status:
Current
Principal investigator:
Professor Jiook Cha
Lead institution:
Seoul National University, Korea (South)

Our overarching goal is to develop data-driven Machine Learning-based predictive modeling for AD. Firstly, we aim, using genetic and health records data, to determine risk and protective factors for neurodegenerative diseases (e.g., AD). Secondly, we aim to develop deep learning algorithms for brain imaging markers that are associated with the suggested factors and predict risk for AD. Thirdly, we will test the predictive validity and generalizability of the initial deep learning model by retraining it for an independent clinical data. In sum, this study will generate novel machine learning methodologies for brain imaging analysis informed by multi-dimensional data. This research meets UK Biobank?s purpose because the target disease?Alzheimer?s disease and dementia?is a significant societal problem and of public interest. Particularly, we aim to develop novel data analytics based on state-of-the-art Artificial Intelligence (e.g., deep convolutional neural network) that has yet to be widely applied to brain imaging data. This development will impact not only Alzheimer?s Disease research but also research of other brain diseases or psychiatric disorders. Data used in Aim #1 includes genetic, electronic health/medical records, emotion/cognition/psychological, and other health-related data (e.g., physical activity, carotid ultrasound, and vision).
Please note that we amended the original application to include the full cohort of participants, without the age exclusion criterion. We are thus requesting data from individuals who have either brain MRI data, genetics, emotion/cognition/psychological data, or miscellaneous data to be used in deep phenotyping for Alzhimer’s disease related dementia as well as other types of dementia.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-human-genetics-to-enhance-discovery-of-genetic-disease-associations

Deep Learning Human Genetics to Enhance Discovery of Genetic Disease Associations

Last updated:
ID:
55964
Start date:
21 April 2020
Project status:
Closed
Principal investigator:
Professor Rick Stevens
Lead institution:
University of Chicago, United States of America

This research applies the latest advances in artificial intelligence, specifically deep learning, to understand how an individual’s genetic code may connect to complex observable characteristics (referred to as phenotypes): for example, the shape and structure of their brains, or how certain individuals may be vulnerable to diseases such as cancer. Our research, if successful, will lead to new computational tools that can recognize early warning signs of a disease or susceptibility to different diseases.

Deep learning (DL) has been particularly successful in biomedical applications; it has been applied to recognize objects in images and videos such as brain tumors and whether patients have Parkinson’s or Alzheimer’s disease, extract phenotypes from electronic health reports, and control a surgical robot’s movement to name just a few.

However, for DL applications to be successful, they need extremely large datasets. We are excited to use the UK Biobank data because of the large number of patients and its intrinsic diversity. We hypothesize that DL approaches can outperform traditional methods, but only when trained on a sufficient number of records. But what exactly is this sample size, and does it vary from disease to disease? Furthermore, what is the nature of inherent bias and vulnerabilities in the context of varying data sample sizes?

We are interested in examining the relationship between sample size and prediction accuracy by phenotype for DL methods. We are also interested in understanding the impact of the complexity of the phenotype in relationship to prediction accuracy, variation and error. We will build tools incrementally, beginning with a tool which uses a patient’s genetic information to make predictions about different phenotypes. We will start with phenotypes associated with the brain, and head and neck cancers. Quite distinct phenotypes with different complexities. This project will be completed in about two years.

The use of DL approaches to characterize complex genotypic-phenotypic associations has been challenging mainly due to the lack of (large, well annotated) datasets. The UK-Biobank datasets offer an unprecedented opportunity to evaluate these new techniques in the context of the questions posed above.

We speculate that DL techniques can have a transformative impact in enabling potentially patient specific treatments based on a variety of prediction tasks, such as the early prediction of Alzheimer’s, Parkinson’s, best drug treatments, and patient outcomes.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-in-cardiac-hemodynamics-enhancing-non-invasive-prediction-with-multimodal-data-and-genetic-insights

Deep Learning in Cardiac Hemodynamics: Enhancing Non-Invasive Prediction with Multimodal Data and Genetic Insights

Last updated:
ID:
595899
Start date:
20 April 2025
Project status:
Current
Principal investigator:
Dr Christoph Reich
Lead institution:
Heidelberg University Hospital, Germany

Cardiac hemodynamics play a crucial role in diagnosing and managing heart failure. Current invasive methods are resource-intensive and pose risks, while non-invasive alternatives often lack reliability. We recently demonstrated that AI-driven analysis of cardiac MRI images could accurately predict LVEDP, offering a non-invasive alternative to catheter-based measurements (Lehmann et al., 2024). By integrating cardiac MRI and genetic data from the UK Biobank, this project seeks to advance the understanding of hemodynamic phenotypes and develop AI-driven diagnostic tools. This project aims to leverage the UK Biobank’s unique dataset with paired cardiac MRI and genotype data to uncover genetic determinants of cardiac hemodynamic parameters and enhance diagnostic prediction. The research focuses on:
– What genetic variants are associated with hemodynamic parameters such as LVEDP, PAWP and SVR?
– What insights can be gained about the interplay between phenotypic and genotypic determinants of cardiac function?
To solve these questions we aim to conduct GWA studies to identify genetic variants linked to hemodynamic traits using UK Biobank’s genotype data. We will begin by applying our AI-LVEDP model, previously developed and validated, to predict LVEDP using cardiac MRI data from the UK Biobank. Following the predictions, we will conduct GWAS to identify genetic variants associated with AI-predicted LVEDP as well as hemodynamic parameters currently in the pipeline. This will enable us to explore genotype-phenotype associations specifically linked to the predicted hemodynamic variables. The same approach will be extended to other hemodynamic parameters once the models are available and validated. Finally, we aim to investigate the clinical and epidemiological relevance of AI-predicted hemodynamic parameters. Specifically, we will assess their prognostic ability for cardiovascular outcomes (cardiovascular death, all-cause mortality, HF hospitalizations).


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-methods-for-early-and-personalized-prediction-of-ascvd-heart-failure-and-cardiometabolic-dysfunction

Deep Learning Methods for Early and Personalized Prediction of ASCVD, Heart Failure, and Cardiometabolic Dysfunction

Last updated:
ID:
1046345
Start date:
14 October 2025
Project status:
Current
Principal investigator:
Mr Arvind Srivastav
Lead institution:
Veevo Technologies, Inc., United States of America

Scientific Rationale: Current cardiovascular and metabolic risk stratification tools are often based on a limited set of risk factors at a single time point. They lack the sensitivity to detect early-stage pathophysiological changes and fail to model the complex, time-varying interactions between genetic predispositions, biomarkers, and lifestyle factors over an individual’s lifespan. The UK Biobank’s rich, longitudinal, and multi-modal dataset provides a unique opportunity to move beyond population-level risk scores toward individualized, dynamic risk prediction models.

Research Questions:
1. Can deep learning models that integrate longitudinal and multimodal health data provide a more accurate, lifelong risk trajectory for Atherosclerotic Cardiovascular Disease (ASCVD) than current standard models?
2. Can cardiac imaging and biomarkers data be used to identify novel, pre-clinical phenotypes of heart failure (HF) risk?
3. What are the early indicators of metabolic dysregulation that can predict incident Type 2 Diabetes (T2D) long before established diagnostic criteria are met?

Objectives:
1. Research and develop deep learning methods for personalized, longitudinal prediction of ASCVD risk using multimodal health data.
2. Identify and model early indicators of HF risk by integrating imaging, genetic, and biochemical data.
3. Research and develop predictive models for near-term T2D risk using early correlates of cardiometabolic dysfunction.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-methods-for-early-detection-of-myocardial-ischemia-and-sudden-cardiac-death-using-digital-ecgs-genomic-and-physical-activity-data

Deep learning methods for early detection of myocardial ischemia and sudden cardiac death using digital ECGs, genomic and physical activity data

Last updated:
ID:
78851
Start date:
10 November 2022
Project status:
Current
Principal investigator:
Dr Xin Li
Lead institution:
University of Leicester, Great Britain

This project aims to assist in the identification of persons at risk of heart attacks and sudden unexpected deaths using ECG, genetic and activity data. ECG data is a non invasive means of assessing the electrical activity of the heart and is used everyday in clinical practice in the NHS, though its interpretation is limited to visual assessment by a trained physicians. Genetic data offers an alternate view point from which we might be able to predict poor health as does the physical activity data from wrist worn devices much like a fitbit. Novel algorithms can learn from large database and identify features that are otherwise hidden, to help identify those at risk of heart attacks and death. The UK biobank has a large resource of ECG, genetic and physical activity data which we believe hold these hidden features which we hope to find using complex computer algorithms. This would allow clinicians to intervene and reduce the risk of heart attacks and death of thousands of patients in the UK annually. We expect the project to take 3 years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-model-to-predict-physiological-ageing-rate

Deep Learning Model to Predict Physiological Ageing Rate

Last updated:
ID:
37212
Start date:
25 October 2019
Project status:
Closed
Principal investigator:
Dr Zichen Wang
Lead institution:
Icahn School of Medicine at Mount Sinai, United States of America

We aim to discover ageing related factors from the multitude of variables profiled in the UK Biobank.

Aim 1. Reapply our Deep Learning methodology to an independent electronic medical records (EMR) dataset. We will employ our original Deep Learning model to predict chronological age as a proxy for physiological age utilizing the biomarker data from the UK Biobank. We will interpret the predictive model by investigating the contribution of biomarkers to the prediction of physiological ageing, and compare these results to our original study.

Aim 2: Perform association study to discover how physiological, clinical and genetic factors affect ageing rate. Ageing is a risk factor for many complex diseases, whereby ageing rate varies across individuals. This research will use the UK Biobank’s data to: 1) Validate and improve our previous physiological ageing predictive model; 2) Identify physiological, clinical and genetic factors that affect physiological ageing rates across individuals. The predictive model will be helpful for indicating general health status for individual patients. All together, the new knowledge gained from this study can inform new opportunities in personalized healthcare. We will use biomarker data from all individuals to build a predictor capable of estimating the physiological age of patients. Such estimated physiological age will enable us to identify individuals with abnormal ageing rates. We will then perform association analyses to identify genetic, clinical and environmental factors affecting the divergent physiological ageing rates for these individuals. The full cohort will be needed for our analysis/project.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-models-for-brain-age-prediction-and-unraveling-patterns-of-aging-with-application-to-neurological-disease-diagnosis

Deep learning models for brain age prediction and unraveling patterns of aging with application to neurological disease diagnosis

Last updated:
ID:
68981
Start date:
8 February 2021
Project status:
Current
Principal investigator:
Professor Ziga Spiclin
Lead institution:
University of Ljubljana, Slovenia

Aging and related structural changes of the brain throughout lifespan have been the subject of research for multiple decades. The goal of this study is to contribute to research of a promising biomarker for neurological health, namely brain age. The expected research is expected to take place during the next 2.5 years. During this time, we wish to develop algorithms capable of prediction of an individuals age, given his or hers MR image of the head. The idea is that this predicted age is close to the chronological age of a healthy individuals. A larger gap may indicate an increased neurological aging and would require further examination. The ultimate aim of the research is not only to develop algorithms, but also to unravel the patterns of brain aging, which can lead to a better understanding of many neurodegenerative diseases, such as Alzheimer’s dementia and multiple sclerosis, and thus hopefully provide a tool for early diagnostics of these diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-models-for-predicting-phenotypic-quantitative-traits-from-high-density-genotyping-data

Deep Learning models for predicting phenotypic quantitative traits from high density genotyping data

Last updated:
ID:
96326
Start date:
29 March 2023
Project status:
Current
Principal investigator:
Dr Eric Barrey
Lead institution:
French National Research Institute for Agriculture, Food and Environment (INRAE), France

Aims
The use of genotyping methods is becoming a routine analysis in predictive medicine (oncology), genealogy research, Gene Wide-Association Study in diseases or psychological traits, and also in another field : animal genetic which is our research topic for production and health. The aims of our project is develop Deep Learning (DL) or Machine Learning (ML) models to predict quantitative phenotypes by using the genotyping data of the individuals.

Scientific rationale
The big amount of genotyping data overreached the computation capacity of the classical methods of phenotype prediction. In order to improve the predictive models of quantitative phenotypes, we propose to apply DL/ML methods more adapted to the big data and non-linear response.

Project
The development of these DL models needs a lot of data to be trained in a first step before being able to predict in a second step. We will implement DL/ML models i) to generate artificial genotypes and phenotypes using generative adversial neural networks model (GAN) and then ii ) to combine real and artificial data to train a DL/ML models to predict quantitative phenotypes by using the genotyping data of the individuals.

Impact
The generative model could be useful to produce artificial data genotypes-phenotypes nearly similar to real data but really anonymous that can be used for other statistical analysis, GWAS or models. The optimisation of these DL/ML models applied to genomics could be a determinant progress for many applications in human medicine and animal genetics.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-of-personal-health-genomics-and-imaging-data-for-early-detection-of-prostate-and-breast-cancers

Deep learning of personal health, genomics, and imaging data for early detection of prostate and breast cancers

Last updated:
ID:
64085
Start date:
10 November 2020
Project status:
Current
Principal investigator:
Dr Jun Deng
Lead institution:
Yale University, United States of America

The aim of this project is to develop a multi-modality cancer risk model based on deep learning of personal health, genomics, and imaging data for early detection of prostate and breast cancers. The scientific rationale for this project is that, current screening methods for prostate and breast cancers have led to substantial over-diagnosis and over-treatment, adding no benefits to millions of cancer patients worldwide while impairing their quality of life. In this setting, the development of a multi-modality cancer risk model will help to both improve the way we stratify and detect these cancers at early stages and identify the relevant risk factors for early prevention of these diseases. This project will last 36 months. Upon completion, this project will provide an accurate, non-invasive, and cost-effective approach for prostate and breast cancer screening to avoid over-diagnosis and over-treatment, as well as advance our understanding of the correlations between these cancers and various cancer risk factors for targeted prevention.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-prediction-of-complex-traits

Deep learning prediction of complex traits

Last updated:
ID:
27081
Start date:
1 April 2017
Project status:
Closed
Principal investigator:
Professor Jacques Fellay
Lead institution:
Ecole Polytechnique Federale de Lausanne (EPFL), Switzerland

The genetic architecture of complex human traits is still largely unclear, with known genetic associations only explaining a fraction of the estimated heritability. There has been no report yet describing the use of artificial intelligence for phenotypic trait prediction. We here aim at developing deep learning algorithms that can predict complex human traits from genome-wide genotyping data, starting with highly heritable but genetically poorly explained phenotypes like height and body mass index. Developing new strategies and tools that will allow the community to extract more information from large-scale genotyping data is a necessary step on the road to more personalized healthcare. We will attempt to train models that can predict complex phenotypes (e.g. height and BMI) from [1] lists of associated polymorphisms previously identified through GWAS approaches, and [2] all common human genetic variation, without a priori selection of variants. We will use different variants of Deep Neural Networks to find and quantify correlations between the SNPs and the phenotypes. Full Cohort


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-research-on-idiopathic-urology-phenomena

Deep learning research on idiopathic urology phenomena

Last updated:
ID:
58180
Start date:
9 March 2020
Project status:
Current
Principal investigator:
Mr Robert James Arbanas
Lead institution:
University of Zagreb, Croatia

The goal of the research is to better understand risks and causes leading to formation of kidney stones. Although many risk factors are already known, for example low fluid intake, or set of medical conditions including genetic determinants, we still don’t fully understand why large percentage of the most common kidney stones form.
By applying modern machine learning (“artificial intelligence”) algorithms on available patient data including basic diagnostics, sociodemographic and lifestyle data our project will try to present kidney stones formation prediction model.
Our goal is to develop information models and put the prediction information early in front of prospective patient and help avoid painful urology procedures related to kidney stones.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-to-predict-mortality-risk-from-ecg-recordings

Deep learning to predict mortality risk from ECG recordings

Last updated:
ID:
85488
Start date:
22 June 2022
Project status:
Current
Principal investigator:
Dr David Michael Farmer
Lead institution:
Massachusetts General Hospital, United States of America

The aim of the study is to explore the use of deep-learning convolutional neural networks (CNNs) for the analysis of electrocardiograms (ECGs) to predict long-term mortality risk.
The electrocardiogram (ECG) is a physiological signal that represents the electrical activity of the heart. The ECG is a tool used in clinical medicine to provide information on the physiological and structural condition of the heart. Although the acquisition of the ECG recording is well standardized and reproducible, the reproducibility of human interpretation of the ECG varies significantly according to levels of experience and expertise. Computer-generated interpretations of the ECG have been used for several years; however, these interpretations are based on predefined rules and manual pattern or feature recognition algorithms that do not always capture the complexity and nuances of the ECG signal. More recently, artificial intelligence (AI) in the form of deep-learning convolutional neural networks (CNNs) has been deployed to analyze routine 12-lead ECG recordings. CNN’s have been successfully developed to detect left ventricular dysfunction, atrial fibrillation, hypertrophic cardiomyopathy, and an individual’s age and sex on the basis of ECG alone. Advanced deep-learning techniques have enabled rapid and precise interpretation of ECG signals, making the ECG a powerful, non-invasive biomarker for diagnostics and cardiac event prediction.
Developing risk models using electrograms can be used to better inform the public of risk and help improve mortality.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-learning-with-model-interpretation-and-uncertainty-quantification-for-polygenic-risk-scores-and-genome-wide-association-studies

Deep learning with model interpretation and uncertainty quantification for polygenic risk scores and genome wide association studies

Last updated:
ID:
169860
Start date:
6 March 2024
Project status:
Current
Principal investigator:
Professor Chongle Pan
Lead institution:
University of Oklahoma, United States of America

Complex traits and diseases are often impacted by one’s genetic composition. And predictive genomics provides a method to gain insight into how genetic variations can be used to estimate genetic risk for complex traits and diseases. Our project aims to enhance the accuracy and reliability of genetic predictions across a range of traits and diseases. We will develop, train, and test machine learning models designed to predict risk for these complex traits and diseases. In addition, we will perform model interpretation to understand why a model predicts the way it does. This will allow for the identification of significant features used in the model’s decision-making process. We also seek to dive deeper to understand and communicate how confident we can be in the predictions through uncertainty quantification techniques. By systematically quantifying uncertainty, we aim to enhance the trustworthiness of genetic risk assessments, critical for informed medical decision-making.

We anticipate this project will last approximately three years from accessing the data to publication of results. The public interest is served through more accurate and reliable genomic predictions, empowering healthcare providers with tools to provide personalized care plans, ultimately improving patient outcomes and advancing the broader understanding of genomics in the public health domain.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-machine-learning-algorithm-for-cancer-risk-prediction-using-genes-environment-and-clinical-data

Deep Machine Learning Algorithm for Cancer Risk Prediction Using Genes, Environment, and Clinical Data

Last updated:
ID:
194370
Start date:
30 April 2024
Project status:
Current
Principal investigator:
Professor Jinluan Li
Lead institution:
Fujian Medical University, China

Our research project aims to make a significant contribution to understanding and predicting the development of cancer. By utilizing advanced techniques such as deep learning and integrating multiple data sources, including genetic, environmental, and clinical data, we expect to develop a powerful model capable of accurately assessing an individual’s risk of developing cancer. This project will take approximately three years, during which we will carefully collect and preprocess the necessary data, develop sophisticated deep learning algorithms, and rigorously evaluate their performance.
The impact of this research on public health could be transformative. With improved risk prediction, healthcare providers can identify high-risk individuals earlier, allowing for targeted interventions and personalized prevention strategies. This has the potential to significantly reduce cancer incidence and mortality. In addition, by exploring the complex interactions between genetic, environmental and clinical factors, we aim to improve our understanding of cancer mechanisms and identify potential new biomarkers or targets for future therapeutic advances.
Overall, this comprehensive and interdisciplinary approach has the potential to revolutionize cancer prevention and treatment. It has the ability to improve public health by informing evidence-based interventions and policies, leading to better patient outcomes and quality of life. In addition, findings from this research project may lay the groundwork for future precision medicine approaches and personalized treatment plans, ultimately reducing the burden of cancer on individuals, families, and society as a whole.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-neural-modular-genetic-networks

Deep neural modular genetic networks

Last updated:
ID:
43117
Start date:
4 February 2019
Project status:
Closed
Principal investigator:
Dr Mika Gustafsson
Lead institution:
Linkoping University, Sweden

Complex diseases are caused by the interaction of many factors, sometimes hundreds of them, as in one of our most interesting complex diseases, multiple sclerosis. These are both genetic, i.e. in the human DNA, and epigenetic, i.e. are influenced by the environment. Many of the factors and their interactions are not yet known. Because of this and other factors, most drugs for complex diseases are effective for less than half of the patients.

A key limitation to understand complex diseases is that the underlying statistical assumptions most often are adaptations of the idea testing only a few factors at the same time for association to the disease. To make most of the emerging big data sets measuring several hundreds of thousands of factors simultaneously we will combine standard tools from artificial intelligence (AI) with our previously successful network medicine concepts that we previously published in well-reputed journals like Science Translational Medicine and Cell Reports.

Artificial intelligence (AI) has recently shown enormous success in applications such as cancer diagnostics, self-driving cars, language translation, and board games. We want to explore whether those methods also could be used to improve our understanding of the human DNA. Our AI application will distil information from DNA and other biological data using deep genetic auto-encoders combined with biological network analysis. This information could increase the efficiency in other scientific studies of complex diseases. We will make the results available to the research community as computational processed drug discovery tools and present our findings to clinical collaborators and suggest potential new biomarkers. Thus, enabling increased precision in disease and drug association studies.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-neural-network-based-analysis-of-resting-state-functional-connectivity

Deep neural network based analysis of resting-state functional connectivity

Last updated:
ID:
66930
Start date:
28 May 2021
Project status:
Current
Principal investigator:
Professor Jae-Jin Kim
Lead institution:
Yonsei University, Korea (South)

Aims : Our research aim is to develop and validate deep learning methods to find the representation of the complex brain networks.

Scientific rationale: The brain communicates between its regions as a complex network to maintain its function. Understanding this complex network property can potentially shed light to reveal the biological underpinnings of human cognition and behavior. Recent deep learning methods are becoming capable of learning representation of complex network structures.

Project duration: The expected duration of the research is 24 months (2 years)

Public health impact: The proposed project is expected have impact on public mental health care. Understanding the biological underpinnings of the complex brain network can potentially guide a new way to the early diagnosis of psychiatric/neurological disorders, such as the Alzheimer’s disease or schizophrenia.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-phenotyping-for-common-and-rare-diseases

Deep phenotyping for common and rare diseases

Last updated:
ID:
107083
Start date:
31 August 2023
Project status:
Current
Principal investigator:
Mr Thore Manuel Buergel
Lead institution:
Pheiron GmbH (i.Gr.), Germany

Our research project aims to bring artificial intelligence (AI) and genomics together to transform how we understand and treat diseases. In the world of medicine, understanding diseases and their underlying causes is crucial for creating effective treatments. This understanding often comes from examining patients’ genetic information and manually curated sets of diagnostic codes from linked health records. However, the current method of doing this is prone to errors and doesn’t take advantage of all the complex data that we can now gather.

In the modern clinical context, a wealth of data beyond traditional health records is available. This includes ‘Omics’ data (proteomics, metabolomics, etc.), which provides a detailed view of a patient’s biological makeup, as well as imaging data (like MRIs and retinal fundus images) and functional test results (like ECGs). But, currently, we don’t have efficient ways to use all this complex data in understanding diseases.

That’s where our research comes in. We aim to develop sophisticated AI methods to draw meaningful conclusions from this wealth of complex data. Using deep learning, we’ll train models to find disease-relevant information from all this data, helping us to develop a more precise and detailed picture of different diseases – we call this ‘deep phenotyping’.

Once these AI models are trained, we’ll examine the extracted data to understand the similarities and differences across various types of data. We’ll then use this refined and combined data to improve our understanding of the genetic architecture of various diseases. The goal is to enable a more nuanced understanding of diseases and their genetic underpinnings.

The project is expected to last for three years. By the end of this period, we aim to have a well-established AI framework capable of harnessing complex biomedical data to produce deep disease phenotypes. These phenotypes will be used to augment the available genetic evidence, enhancing our understanding of diseases and potentially opening up new avenues in drug target discovery.

The potential impact on public health is substantial. By enhancing our understanding of disease mechanisms, our research could revolutionize the way diseases are characterized and treated. This could lead to more accurate diagnoses, more effective treatments, and ultimately, improved health outcomes for patients. Furthermore, our project could also pave the way for a more personalized approach to healthcare, with interventions tailored to the specific disease profiles of individual patients


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-phenotyping-of-major-depressive-disorder-in-adults

Deep Phenotyping of Major Depressive Disorder in Adults

Last updated:
ID:
53246
Start date:
9 March 2020
Project status:
Current
Principal investigator:
Professor Andrea Cipriani
Lead institution:
University of Oxford, Great Britain

Depression is a very common major health problem. With 350 million people affected in the world, depressive disorder is the second leading cause of global burden. The high direct and indirect costs for major depression are largely due to significant problems in treatment. There are several effective treatments for depressive disorder, but the key challenge is how best to use and deliver currently available effective treatments.
The UK Biobank is a National Health Service data archive, which contains a variety of medical information including genetic, brain imaging, biomarkers, cognitive function and reported medical histories for participants with and without major depressive disorder. These data offer the opportunity to investigate in a large cohort of individuals a great number of variables possibly related to depression and to treatment response.
Our aim is to perform a deep phenotyping of major depressive disorder in adults. We will also link clinical information available in medical records on the UK-CRIS dataset with the UK Biobank data to determine antidepressant response trajectories and associated clinical prognostic factors. It will help us to understand better prognostic factors and determinants of antidepressant response in people with major depressive disorders and will ultimately lead to improvement to our clinical practice.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-phenotyping-of-patients-with-a-range-of-chronic-pain-conditions-in-the-uk-biobank-using-a-machine-learning-approach

Deep phenotyping of patients with a range of chronic pain conditions in the UK Biobank using a machine-learning approach.

Last updated:
ID:
45465
Start date:
29 July 2019
Project status:
Current
Principal investigator:
Dr Andrew Segerdahl
Lead institution:
University of Oxford, Great Britain

Chronic pain is a common and disabling problem which affects up to half of adults in the UK. The current treatments for chronic pain are often not very effective. This is partly because we do not fully understand how the pain is generated. We know that part of the problem is due to how pain signals are processed in the brain. By doing brain scans on patients with chronic pain, we can see how the different areas of the brain react to pain and how they are connected to one another. Studies involving brain scans in patients with chronic pain usually only involve a small number of patients. This means that we cannot be completely sure that what has been found so far is definitely correct. In addition we might have missed some important differences between patients that might exist even if they have been diagnosed with the same painful condition.

The UK biobank study aims to image 100,000 volunteers by 2020. Of these participants, a significant percentage are likely to have one of the painful conditions that we would like to study. This means that we will be able to conduct a very powerful study to investigate the how the brain reacts to pain. We will also use a technique called ‘machine-learning’ to help analyse the data. This approach uses computer programming to find new patterns in the data which we may not otherwise be aware of. In doing so, we may uncover a better understanding of how the brain is working in people with chronic pain and we hope that this will help to shape future research looking at the best ways to help improve the quality of life for these people.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-phenotyping-of-ugt-human-knockouts

Deep phenotyping of UGT human knockouts

Last updated:
ID:
398221
Start date:
11 December 2024
Project status:
Current
Principal investigator:
Professor Chantal Guillemette
Lead institution:
Universite Laval, Canada

Glycosyltransferases (UGT) are metabolic enzymes that regulate the biological action and the elimination of drugs and of multiple endogenous metabolites to keep them in balance. Among the UGT family of 22 enzymes that have diverse roles in nearly all body organs, UGT2B17 and UGT2B28 are frequently completely deleted from the human genome. In other words, individuals without any of those genes are naturally and completely deficient (or knockout, KO) in UGT2B17 or UGT2B28 or both enzymes. Despite the relatively frequent occurrence of these KOs in the population (10% and 3% of the Caucasian population are UGT2B17 KO and UGT2B28 KO, respectively) and the growing evidence of their association with diseases including some cancers, the metabolic and health-related consequences of not having one of those genes have not been comprehensively determined. Our recent study of the Canadian Longitudinal Study on Aging revealed important differences in the systemic metabololomic profiles (small molecules measured in blood) of UGT KO individuals, with a strong sexual dimorphism. In addition, multiple associations of changed metabolites with diverse common metabolic diseases such as diabetes and hypertension as well as with arthritis and osteoporosis were found and require validation.

Our objective is to leverage on the UK biobank to validate and expand our initial findings supporting their significant impact on health and disease. Our research project is initially planned for 3 years but is expected to be prolonged if promising first results are obtained. Our objective is to gain a better understanding of the impact of a UGT deficiency on metabolite and protein blood profiles, and how it impacts health and disease. By identifying the metabolites and the proteins affected by the absence of these genes, we will greatly improve our understanding of the functions of these UGT enzymes in the human body and potentially identify strategies to mitigate their detrimental effects or enhance their beneficial impacts. This project will thus lay the groundwork to understand the impact of two of the most frequently deleted gene of the human genome.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deep-unsupervised-clustering-of-combined-multi-modal-neuroimages-and-clinical-data-to-improve-subtyping-of-population-wide-mental-and-behavioural-disorders

Deep unsupervised clustering of combined multi-modal neuroimages and clinical data to improve subtyping of population-wide mental and behavioural disorders.

Last updated:
ID:
79105
Start date:
6 July 2022
Project status:
Closed
Principal investigator:
Mr Jorge Garcia Condado
Lead institution:
BioCruces Health Research Institute, Spain

The aim of this project is to group subjects in the UK Biobank neuroimaging cohort according to characteristics in the images taken of the brain of each subject. Several different types of brain images were taken of each patient. Using computer algorithms we want to leverage this richness of information to separate subjects into groups according to how their brains look in each imaging modality.

After we have separated subjects into different groups we will see what characteristics subjects in a group share and how they are different to subjects in other groups. We will focus both on characteristics from their brain images (e.g. people in a group might have larger brain structures) and on clinically relevant data (e.g. people in a group might have all been diagnosed with Alzeihmer’s disease). Finally, genetic analysis will be carried out to see if in each group genetics plays a role.

Most mental and behavioural disorders typically share intersecting symptoms making it challenging to perform a precise categorization. It is important to do distinguish diseases and look at each disease as possibly having different symptoms in different patients. Therefore by looking at groups of subjects that share similar characteristics we can help clinicians better identify patients that might share similar disease progression. This can only be done nowadays because large amounts of data are required for these types of grouping strategies and these were not available until initiatives like the UK Biobank were carried out.

This project is part of a PhD thesis expected to last 3 years (36 months). The first part of the project will last about 2 years and will focus on grouping patients together that share similar characteristics according to neuroimaging data. Once these groups have been determined the second part of the project will involve consulting with clinicians on how to use the information extracted to characterise each group.

The prevalence of mental and behavioural disorders is only increasing. For example, the two most prevalent neurological disorders, Alzheimer’s and Parkinson’s disease already affect over a million people just in the UK. As popoluation’s age the prevalence of such diseases is only going to increase. Mental and behavioural disorders are not only terrible for the patient but can have an impact in the life of relatives and friends. Therefore being able to diagnose them early is crucial to help all involved better understand the situation.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/define-disease-endotypes-agnostic-of-conventional-diagnostic-classes

Define disease endotypes agnostic of conventional diagnostic classes

Last updated:
ID:
43138
Start date:
10 September 2018
Project status:
Current
Principal investigator:
Dr Delphine Marion Rolando
Lead institution:
BenevolentAI Bio Ltd, Great Britain

Many drugs that are approved to treat everybody with a certain disease diagnosis, may work well for some people with the diagnosis while providing no benefit for others. One of the reasons for this is that people with the same diagnosis of disease, although they may have similar seeming symptoms or signs, may not have the same cause of the diseases at the level of molecular or cellular changes. This may be because there a several molecular/cellular routes to similar manifestations of disease. This mismatch between diagnosis and molecular/cellular cause, results not only in lack of predictable benefit from marketed drugs but also in difficulty in testing the efficacy of experimental drugs. Additionally this results in some molecular causes never being treated, because they do not fall neatly within a diagnosis.

Here we will attempt, using machine learning, to define disease not by diagnostic codes but by empirical measures such as genotype, biomedical imaging or by more granular symptom self-reporting. We hypothesise that groups of participants with similar molecular causes of disease can be discerned within the UK Biobank cohort. We plan to define these UK biobank participant groups and then compare them to the diagnoses the participants have received. We will compare different ways of building the groups (from DNA sequence alone, or from image or clinical features as well) with how predictive they are of diagnoses. We will also compare what we learn in UK biobank with other ways of subclassifying disease. We will publish these findings and also use them to inform the drug discovery work within BenevolentAI, a UK based biotechnology company.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/defining-a-novel-disease-entity-and-unraveling-disease-mechanisms-to-improve-drug-development-efficiency-using-uk-biobank-data

Defining a Novel Disease Entity and Unraveling Disease Mechanisms to Improve Drug Development Efficiency Using UK Biobank Data

Last updated:
ID:
784335
Start date:
25 September 2025
Project status:
Current
Principal investigator:
Dr Seunghwan Jung
Lead institution:
Hanmi Pharmaceutical Co., Ltd, Korea (South)

Understanding diseases is crucial for improving quality of life through effective treatments. Advances in biological analysis allow multifaceted disease studies using imaging, multi-omics, and clinical data. Such integrative approaches redefine complex diseases, like cancers, metabolic diseases, autoimmune diseases, and aging, as modifiable conditions. However, both remain challenging due to their complexity and unclear definitions. Moreover, integrating vast data into uncover disease mechanisms is a formidable task.
To tackle this, we propose a stepwise research approach using UK Biobank data. First, we aim to develop aging prediction models with multi-omics data, identifying key biological markers. Second, we aim to define a novel disease entity, similar to metabolic syndrome, that can serve as a marker for cardiac disease, diabetes, and related conditions. This definition will establish diagnostic criteria, providing a standardized framework for identifying individuals at risk. Third, we will apply machine learning to multi-omics data to elucidate disease-gene relationships, enhancing insights into complex diseases. Finally, with refined disease definitions and identified pathways, we will optimize drug mechanism studies, identify new indications for existing drugs, and discover novel targets, improving drug development efficiency for public benefit.
Our research will contribute to precision medicine by advancing disease prediction and mechanism insights. Integrating multi-omics and machine learning will deepen biological understanding, enabling targeted therapies. Our models will comply with UK Biobank’s AI policy, anonymizing data. Derived variables from the AI model will be returned to UK Biobank for other researcher’s access. We will also disseminate our research findings in academic journals or conferences. Utilizing UK Biobank, we aim to bridge data-driven discovery and translational medicine, fostering public health and pharmaceutical innovation.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/defining-a-signature-of-prodromal-parkinsons-disease-in-the-uk-biobank-cohort

Defining a signature of prodromal Parkinson’s disease in the UK Biobank cohort

Last updated:
ID:
95506
Start date:
16 December 2022
Project status:
Current
Principal investigator:
Professor Michele Tao-Ming Hu
Lead institution:
University of Oxford, Great Britain

We currently have no cure or treatments that slow down disease progression in Parkinson’s disease (PD), which will double in prevalence in the next 25 years, affecting >10 million people worldwide. This relentlessly progressive condition has devastating consequences for individuals and families, being a leading global cause of movement-related disability and dementia.

There is compelling evidence for a causal relationship between sleep disturbance and disorders such as PD and dementia. However, it isn’t clear how one causes the other. Medical interventions can normalize sleep and may therefore represent a novel strategy to slow down disease progression or delay the onset of Parkinson’s (PD). It is therefore critical to determine how sleep problems may accelerate neuronal death and its underlying biological pathways.

Patients with a disorder called REM-sleep behaviour disorder (RBD) represent an emerging group at high risk of future PD. These individuals suffer a sleep disorder occurring in the dream phase of sleep, the rapid eye movement (REM) phase. Subjects with so called REM-sleep behaviour disorder (RBD) wake their bed partners during violent and vivid dreams, which they enact through movement and vocalization, e.g. kicking their bedpartner. The Gold-standard diagnosis of RBD is usually made using a technique called polysomnography (PSG), recording bodily functions including brain activity, eye movements, muscle activity, heart rhythm, blood pressure, and body movement during sleep.

Research studies have shown that 6 % of RBD patients convert to PD each year or a related neurodegenerative condition e.g., dementia. In addition, PD patients with symptoms of RBD manifest higher rates of dementia, cognitive, and motor symptom progression.

RBD patient cohorts therefore represent an extraordinary opportunity to capture the
early molecular/cellular mechanisms preceding the onset of PD symptoms, evident
only after >50% of dopaminergic neurons in the brain have been lost. However, so far, no molecular or cellular mechanism has been identified explaining how an individual converts from the sleep disorder RBD to the neurodegenerative disorder PD.

Our research aims at studying those RBD patients who have not yet developed PD, so that we can capture and characterise the clinical symptoms and underlying mechanisms driving future PD risk. We aim to compare findings from the Discovery sleep-clinic cohort of RBD patients with those from UK Biobank, so we know how common RBD is in the general population and its effect on future PD risk.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/defining-and-redefining-human-disease-at-scale-an-atlas-of-the-human-phenome

Defining and redefining human disease at scale: an atlas of the human phenome.

Last updated:
ID:
58356
Start date:
12 June 2020
Project status:
Current
Principal investigator:
Spiros Denaxas
Lead institution:
University College London, Great Britain

Our understadning of human disease and the different factors which influence our health changes all the time through but the manner in which we define diseases is still based on what clinicians can directly observe. As a result, we have a one-size-fits-all medication treatment for many diseases which does not benefit all patients as they might have the same disease but have significant differences in their genetic material which influenced if the treatment will work or how well it will work. The aims of this project is to use analytical approaches in order to identify and describe how the same disease can vary across different patients. To do so, we will use data from many different aspects of human health available in the UK Biobank, from genetic data and blood data to phenotypic data that get collected when we interact with the healthcare system. The result of this study (which will last 36 months) will improve human health and healthcare by enabling clinicians to accurately identify who will benefit from what drug and by providing insights into the creation of better drugs for patients who do not currently benefit from existing treatments.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/defining-disease-risk-with-genotype-and-phenotype-integration-using-machine-learning-methods

Defining disease risk with genotype and phenotype integration using machine learning methods

Last updated:
ID:
31984
Start date:
22 February 2018
Project status:
Closed
Principal investigator:
Mr Ari Karason
Lead institution:
Genuity Science, Inc, United States of America

The primary aim of this proposal is to use the UK Biobank data to determine whether artificial intelligence methods based on combining genetic risk variants and phenotypic data improve disease risk classification vs. the conventional method of combining common genetic risk variants using a multiplicative model to create polygenic disease risk. This method may demonstrate that there is more information available in large genomic and medical databases such as the UK Biobank that may then be extracted using traditional statistical methods.
The successful outcome of the research proposed in this application will help to improve the calculation of common disease risk, which will facilitate the prevention of diseases and morbidity.

In addition to providing risk insights, our results have the potential to open new avenues of research for disease intervention and overall health.
Application of artificial intelligence (AI) provides another angle for large data analysis. While this newer analysis method has great potential, it requires testing on considerably large and well-curated disease data collections such as the UK Biobank. The breadth of the phenotypic and genotypic data in the UK Biobank will allow us to test the hypothesis that AI methods perform better at defining disease risk. Our study will first combine the different data types using machine learning to determine important risk factors on a large population subset. We will then confirm these results using a different subset.
We would like to include the full cohort for this study.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/defining-factors-that-modify-the-association-between-diabetes-mellitus-and-cardiovascular-disease

Defining factors that modify the association between diabetes mellitus and cardiovascular disease

Last updated:
ID:
96958
Start date:
26 January 2023
Project status:
Current
Principal investigator:
Dr Richard Cubbon
Lead institution:
University of Leeds, Great Britain

Diabetes mellitus (DM) is a condition where blood sugar levels are too high because of reduced action or production of the hormone insulin. Soon, one in ten adults in the UK are expected to have DM. People with DM experience twice the risk of important cardiovascular problems like heart attack, stroke or heart failure, and these problems occur 15 years earlier than in people without diabetes. These risks can be reduced by health lifestyles, medications and other treatments, but even when used optimally, these approaches only modestly reduce the risk of cardiovascular disease in people with DM. We think that this might be because diabetes is often associated with many other health problems that act in conjunction with diabetes to substantially increase the development of cardiovascular disease. UK Biobank provides an excellent opportunity for us to test this premise.

We will test a wide range of ‘modifying factors’ that might act together with diabetes to increase the risk of cardiovascular disease; broadly, these will include other diseases (e.g. kidney disease), other factors in the environment (e.g. air pollution) and inherited variations in genes (e.g. which can lead to inherited heart diseases). We will use a wide range of UK Biobank data to define people with cardiovascular damage or disease, ranging from mild asymptomatic problems at recruitment, through to those who die as a result of cardiovascular problems. Our experiments will test whether people with diabetes experience a greater risk of cardiovascular problems linked with a ‘modifying factor’ than people without diabetes. When we identify ‘modifying factors’, we will aim to understand how these might act together with diabetes to cause cardiovascular problems. We will also look for common features of ‘modifying factors’ that we identify.

The project will span at least 3 years, and will form part of a doctor in training’s PhD project. We hope that our findings will improve the understanding of how diabetes results in accelerated cardiovascular disease, and that this knowledge will eventually lead to more effective preventative therapies. This has to potential to improve the lives of people with diabetes, along with having important implications for healthcare systems and society.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/defining-novel-measures-of-left-ventricular-contractility-and-their-association-with-the-risk-of-incident-cardiovascular-disease

Defining novel measures of left ventricular contractility and their association with the risk of incident cardiovascular disease.

Last updated:
ID:
106965
Start date:
27 September 2023
Project status:
Current
Principal investigator:
Dr Sam Straw
Lead institution:
University of Leeds, Great Britain

Chronic heart failure (CHF) is a common condition characterised by symptoms of breathlessness, fatigue, and fluid retention. CHF also reduces life expectancy and is associated with increased risk of hospitalisation. CHF is currently classified according to left ventricular ejection fraction (LVEF) – the percentage of blood pumped by the heart during each heartbeat. LVEF is used to determine which patients may benefit from medications to improve symptoms and survival, as those with ‘normal’ heart function do not appear to derive benefit from these therapies. We have shown that this method of to assessing heart function does not reliably predict the future risk of adverse outcomes, especially amongst those who have ‘normal’ heart function. However, when using novel measures of heart function which incorporate the ‘loading conditions’ (the force the heart has to work against) this better reflects future risk and identifies around one third of patients with ‘normal’ heart function who might stand to benefit from receiving medications currently only given to those with impaired heart function. We wish to understand the normal values for cardiac contractility in a healthy population. We will then compare the measurement of heart contractility to LVEF in how it identifies different populations, and how these measures predict the development of cardiovascular disease in the future.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/defining-patterns-of-comorbidity-accrual-before-and-after-the-diagnosis-of-heart-failure

Defining patterns of comorbidity accrual before and after the diagnosis of heart failure

Last updated:
ID:
117090
Start date:
21 November 2023
Project status:
Current
Principal investigator:
Dr Richard Cubbon
Lead institution:
University of Leeds, Great Britain

Heart failure (HF) is a condition where the heart is unable to pump enough to meet the needs of the body, which leads to fatigue, breathlessness on exertion, and reduced life expectancy. People with HF often have many other health problems (or ‘comorbidities’), which can develop before and after HF is diagnosed. The greater the number of these comorbidities a person with HF has, the poorer their quality of life and long-term survival. Currently, our knowledge is very limited regarding how these comorbidities develop over the lifetime of someone with HF. UK Biobank is an excellent resource to study this as it includes many thousands of people with heart failure, along with detailed information about when they developed other health problems, their wider characteristics (e.g. sex, ethnicity), and their long-term survival. As part of a student project (Masters by Research), we plan to study the pattern and timing of comorbidity development in relation to when people are first diagnosed with heart failure. This will help us to find the commonest patterns of comorbidity development before and after HF, along with learning about the outcomes of these people in these groups. This information will help us to identify groups of people where we might one day aim to intervene to slow or prevent comorbidity development, with a view to improving their quality and quantity of life.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/defining-phenotypes-in-diabetic-kidney-disease

Defining Phenotypes in Diabetic Kidney Disease.

Last updated:
ID:
103620
Start date:
31 August 2023
Project status:
Current
Principal investigator:
Dr Rakesh Dattani
Lead institution:
Imperial College London, Great Britain

The number of people world wide with Type 2 Diabetes is increasing, with approximately 700 million people expected to have Type 2 Diabetes by 2045. Type 2 diabetes is associated with a large number of associated healthcare problems related to the effect of long term poorly controlled blood sugars on different body organs including the eyes, heart and kidney. Type 2 Diabetes is the commonest cause of kidney disease worldwide, with a large number of patients subseqeuntly developing kidney failure and a larger number subsequently developing heart disease. It is now recognised that Diabetes related Kidney Disease affects different groups of patients at different speeds and in different ways. It is now also accepted that Diabetes related Kidney disease may consist of different sub-groups which may require different treatment approaches. In identifying these different groups, we should be able to identify personalised targeted therapies for people with this potentially devastating disease. We have previously identified 4 different clinical subgroups amongst patients with long term kidney disease related to diabetes as well as other causes. In this study, utilising clinical data from the UK biobank, we aim to validate the results of our published preliminary study, with the overall aim remaining to determine whether clinical variables may identify different subgroups in patients with Diabetic Kidney Disease, to facilitate further study of underlying mechanisms of diabetes related kidney disease and the development of personalised targeted therapies for people with this potentially devastating disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/defining-physical-activity-phenotypes-based-on-objective-measures-and-dissecting-the-genetic-and-phenotypic-relationships-of-physical-activity-with-diseases-and-health-outcomes

Defining physical activity phenotypes based on objective measures and dissecting the genetic and phenotypic relationships of physical activity with diseases and health outcomes

Last updated:
ID:
29900
Start date:
1 August 2017
Project status:
Current
Principal investigator:
Professor Hongyu Zhao
Lead institution:
Yale University, United States of America

Insufficient physical activity as a major public health concern is associated with increased disease risks and mortality. However, little is known about the genetic predispositions of physical activity and its relationships with diseases, partly due to absence of population-based cohorts with both genetic and phenotypic information. This project will define physical activity phenotypes based on accelerometry data, identify associated genetic components, and further examine the shared genetic and phenotypic architectures between physical activity and many human complex traits, such as obesity, type 2 diabetes, cardiovascular diseases and cancers, to better understand the role of physical activity in disease etiology. This project will help researchers understand how to effectively utilize accelerometry data in physical activity studies. In addition, our results will provide fundamental insights into the genetic basis of physical activity and the complex genetic and phenotypic relationships between physical activity and a spectrum of diseases. Our proposed study will aid in developing effective public intervention strategies to improve population health. We will develop statistical and computational methods to extract informative metrics from accelerometry data and define phenotypes that best characterize physical activity. Then, we employ genetic data from UK Biobank to study the genetic basis of physical activity. Further, by integrating genetic data and phenotypic traits (e.g. from questionnaires, clinical examinations, and medical records), we study the genetic and phenotypic associations between physical activity and a variety of health outcomes. Since a major goal of this study is to systematically investigate the relationships between physical activity and a variety of complex diseases, we would like to request the full UK Biobank cohort subjects (n=500,000).


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/defining-the-association-of-human-endogenous-retroviruses-hervs-and-genes-implicated-in-the-inherited-cardiac-diseases

Defining the Association of Human Endogenous Retroviruses (HERVs) and genes implicated in the inherited cardiac diseases

Last updated:
ID:
93796
Start date:
19 April 2023
Project status:
Current
Principal investigator:
Professor Rameen Shakur
Lead institution:
University of Brighton, Great Britain

Human Endogenous Retroviruses (HERVs) are ‘genomic scars’ whereby viruses have integrated into the human genome throughout our evolution and our interaction with the environment we reside. This form of integration has been occurring into the human genome some 30-40 million years ago and now makes up 8% of the genome. Evidence suggests HERVs may cause a number of neurological and also autoimmune diseases, such as rheumatoid arthritis and, among other rheumatic diseases. However, we are not so clear on the impact of such HERVs which are seen in different populations and are inherited across different subpopulations have on complex diseases such as cardiovascular diseases. To this end we are keen to understand in inherited heart muscle diseases called the Cardiomyopathies could the HERV signatures across patient groups explain why some patients develop disease outputs from the underlying genetic changes in their genomes and others do not even though they may have inherited the same genetic changes. Furthermore, could such markers in ones genome be guide for early signs of disease developing? We have identified potential candidate HERV regions that are having gene expression effects in human diseases, namely in cardiac development and in the instigation of cardiac diseases. This has been achieved through a novel data pipeline we have developed and been able to curate and integrate global data on previously sequenced human normal and diseased patient hearts. Given the variable penetrance in patient genotype and phenotype data in the inherited cardiomyopathies, this HERV map will enable us to understand why some phenotypes seem less clinical homogenous in populations where the genotype maybe prevalent. For example the hypertrophic cardiomyopathies are prevalent in 1 in 200 of the population, but the incidence of clinical cardiomyopathy is not as high. The concept of a trigger or an initial environmental initiator remains, but this could be explained through the differing HERV backgrounds that are seen in individuals, which we know changes and degrades from generation to generation. This has not been undertaken before nor has this novel concept been applied previously in the chronic disease context nor in the complex diseases such as cardiovascular diseases before.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/defining-the-genetic-and-non-genetic-determinants-of-fibrosis

Defining the genetic and non-genetic determinants of fibrosis

Last updated:
ID:
77050
Start date:
26 April 2022
Project status:
Current
Principal investigator:
Professor Louise Wain
Lead institution:
University of Leicester, Great Britain

Scarring of the internal organs occurs in many common diseases, including diabetes (scarring in the pancreas), hypertension (blood vessels), chronic kidney disease (kidney), cirrhosis (liver) and interstitial lung disease (lungs). Scarring of the internal organs can alter the function of these organs dramatically and accounts for one third of all deaths world-wide. People may be affected by more than one scarring disease.
Factors that are believed to contribute to scarring include smoking, alcohol, obesity and infectious diseases such as COVID-19. There are also a number of genetic risk factors that can run in families and can determine the time of onset of scarring in different organs.
It is likely that a unique combination of genetic and environmental risk factors leads to scarring of different organs happening at different times. If we can identify patterns of scarring in early life, we might be able to prevent the development of more extensive scarring in multiple organs in later life, by encouraging people to change their lifestyle or by treating them with medicines.
The aim of our research is to identify why fibrosis occurs in different organs, often within the same patient, and how we can treat it.
Using UK Biobank, we will assess whether scarring which takes place in different organs, involves the same or similar genetic and environmental causes, and will identify the biological pathways that lead to scarring getting worse.
In this way we hope to be able to stop scarring from destroying the organ in which it is found and prevent it spreading to other organs by using a single treatment for scarring, rather than always having to use individual treatments for each organ. These treatments could involve lifestyle changes, such as weight loss and exercise, or new drug treatments. We hope this will improve the quality of life for people with organ scarring and reduce the number of treatments needed to prevent scarring getting worse.
Ultimately, we hope that by understanding the causes of scarring and reasons that scarring gets worse we will prevent the long terms problems that are observed when people get scarring in one or multiple organs in the body.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/defining-the-normal-ranges-of-blood-count-as-a-function-of-demographic-variables-and-genetic-analysis-of-blood-count-parameters

Defining the normal ranges of blood count as a function of demographic variables and genetic analysis of blood count parameters

Last updated:
ID:
13745
Start date:
1 June 2013
Project status:
Closed
Principal investigator:
Dr William Astle
Lead institution:
University of Cambridge, Great Britain

Our groups investigate the genetics and epidemiology of blood counts with the aims of i) optimising blood donation policies and ii) understanding the molecular mechanisms of blood formation. Our ultimate goals are to develop personalized blood donation schedules and to improve diagnosis and therapies for blood diseases.

We have completed genome wide association analyses involving 150,000 individuals and found 140 regions of the genome implicated in regulating the formation of red cells and platelets. We are currently sequencing these regions of the genome in 2000 individuals with extreme red cell volume or platelet count ascertained from the LifeLines cohort of 100,000 individuals, to discover rare variants that affect the formation of these cells.

We seek access to the complete Biobank data to: 1) better define ranges of blood parameters in the UK population, and with unprecedented power, establish how demographic variables such as age and gender affect them. This will be of particular interest for our INTERVAL trial of 50,000 blood donors, which aims to optimise blood donation frequency, by allowing us to better interpret the effects of blood donation. 2) Assess the impact on blood parameters of other variables collected by Biobank, particularly laboratory and nutrition-related data. 3) Investigate blood parameters measured by Biobank that are not routinely measured clinically but that may be better reporters of blood formation such as immature reticulocyte fraction. 3) Study the relationship between blood indices, mortality and cancer. 4) Carry out genome wide association studies, once UKBiobank genotypes become available, of blood parameters to identify genetic determinants of blood formation.

Because of the large number of parameters we wish to study we seek access to Biobank?s complete data on the full cohort but do not need samples. As they become available we also seek access to genotypes, mortality outcomes, repeat measurements and biochemistry results.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/defining-the-preclinical-phase-of-inflammatory-bowel-disease-predict-and-prevent

Defining the Preclinical Phase of Inflammatory Bowel Disease- Predict and Prevent

Last updated:
ID:
953947
Start date:
18 August 2025
Project status:
Current
Principal investigator:
Professor Sun-Ho Lee
Lead institution:
Sinai Health System., Canada

The overarching goal of this project is to define and validate blood-based biomarkers that precede either the onset of Crohn’s disease (CD) or its post-operative recurrence (POR), enabling early risk prediction, mechanistic insight, and targeted prevention. We hypothesize that CD has a prolonged preclinical phase marked by molecular perturbations detectable in asymptomatic individuals before clinical disease develops or re-emerges after surgery.

Research Questions:
– What blood-based proteomic, metabolomic, glycomic, and antibody signatures predict future CD onset and POR?
– Can multi-omics integration uncover shared mechanistic pathways that trigger CD initiation or recurrence?
– When do these biomarkers emerge in high-risk individuals?

Objectives:
– Identify and validate pre-disease and pre-recurrence biomarkers using well-characterized cohorts, including the GEM cohort and a post-operative CD cohort.
– Integrate multi-omics datasets to identify convergent pathogenic networks underlying CD initiation and recurrence.

Scientific Rationale:
CD is a chronic inflammatory condition with complex pathogenesis. Most research focuses on established disease, missing the critical early window when pathogenic mechanisms emerge. Leveraging >2,000 pre-diagnostic blood samples across multiple cohorts, this project uses harmonized multi-omics platforms and integrative analyses to detect early biomarkers and define the sequence of molecular events preceding CD onset and POR. These findings will inform risk stratification, identify therapeutic targets, and lay the groundwork for precision prevention strategies in CD.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/defining-the-role-of-crybg1-germline-alterations-on-tumor-incidence-and-progression

Defining the role of CRYBG1 germline alterations on tumor incidence and progression

Last updated:
ID:
59494
Start date:
4 May 2020
Project status:
Current
Principal investigator:
Dr Michael Christoph Haffner
Lead institution:
Fred Hutchinson Cancer Center, United States of America

Cancer’s most feared feature is its ability to invade into benign tissues and spread to distant sites. Every cell has a structural scaffold, a skeleton termed the cytoskeleton. It consists of a complex interlinked network of proteins that supports the cell and helps to maintain its shape. To invade other tissues, cancer cells need to alter their cytoskeletal properties to become malleable enough to change their shape and to move through tissues. We have recently unmasked an important protein, named CRYBG1 that regulates the cytoskeleton in benign cells and is dysfunctional in cancer. When CRYBG1 is present, the cells’ scaffolding keeps it rigid and correct shape. When CRYBG1 function is lost, cells can remodel their cytoskeleton more frequently change their shape and become capable of invading and migrating to distant locations. Importantly, we have found that the CRYBG1 gene is frequently disrupted in the germline (the DNA sequence shared by all cells in the body) in patients who develop very aggressive forms of metastatic prostate cancer. We therefore hypothesize that genetic alterations in CRYBG1 give cancer cells an early advantage to change shape, migrate, invade and to spread to different tissues.
In this study we aim to examine if these germline genomic alterations in CRYBG1 are present in patients with cancer and if the presence of CRYBG1 mutations is associated with aggressive disease. This information will provide the basis for a novel genetic test for aggressive forms of cancer. Ultimately, we hope that this research will improve the care and survival of patients suffering from cancer and their families.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/degenerative-protein-modifications-dpms-as-molecular-mediators-connecting-socio-demographic-profile-to-the-onset-of-age-related-diseases

Degenerative protein modifications (DPMs) as molecular mediators connecting socio-demographic profile to the onset of age-related diseases

Last updated:
ID:
495589
Start date:
4 February 2025
Project status:
Current
Principal investigator:
Professor Newman Sze
Lead institution:
Brock University, Canada

We have identified a set of degenerative protein modifications (DPMs) in patients’ tissues, resulting from spontaneous chemical reactions like oxidation, glycation, and deamidation. These DPMs play crucial roles in human health and disease. We hypothesize that an individual’s socio-demographic profile interacts with their genome, epigenome, proteome, and metabolome, leading to the formation of harmful DPMs. Consequently, DPMs act as molecular mediators connecting socio-demographic profiles to age-related diseases. We have also identified candidate markers including genes, epigenetic targets, and blood metabolites associated with DPMs accumulation in tissues. In this project, we aim to leverage UK biobank data, along with genomics, epigenomics, and metabolomics datasets. Our objective is to investigate the association of the candidate markers with the clinical outcomes


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/delineating-germline-genes-that-predispose-to-cancer

Delineating Germline Genes that Predispose to Cancer

Last updated:
ID:
50092
Start date:
29 November 2019
Project status:
Current
Principal investigator:
Professor Vijai Joseph
Lead institution:
Memorial Sloan Kettering Cancer Center, United States of America

We know of some genes that cause high risk of cancers, such as BRCA1/2. Other genes likely exist that also increase such risk, but we have not found them all. This proposal looks at the baseline risk for those genes in cancer-unaffected individuals and then tests them against specific cancer groups. We have programs that can find the bad variants quickly across all samples. Our research will give an overview of such cancer-causing variant burden and provide a landscape that depicts risks due to specific genes in different cancer types.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/delineating-the-relationship-between-human-evolutionary-history-on-the-architectural-complexity-and-predictability-of-traits

Delineating the relationship between human evolutionary history on the architectural complexity and predictability of traits

Last updated:
ID:
104628
Start date:
18 August 2023
Project status:
Current
Principal investigator:
Dr Joshua Michael Akey
Lead institution:
Princeton University, United States of America

Differences in DNA sequences among individuals contribute to inherited variation in traits and diseases. In addition, patterns of DNA sequence variation that exist among individuals today were shaped by our past. Therefore, a better understanding of human history will facilitate a better understanding of how and why differences in DNA sequences among individuals leads to differences in traits and disease susceptibility. The UK Biobank provides a unique opportunity to ask fundamentally important questions about the relationship between human history and our current distribution and burden of disease. Our research will develop new tools for leveraging the massively large set of DNA sequences in the UK Biobank to infer aspects of human evolutionary history and how our past shapes patterns of heritable variation in traits and disease among contemporary individuals. We will also use this deeper understanding of how human history affects patterns of DNA sequence variation across the genome to develop new tools for decoding the genetic basis of traits and diseases. For instance, some mutations might influence multiple traits or diseases and therefore methods that simultaneously consider two or more traits or diseases would potentially make it easier to identify such mutations. We anticipate the project will take three years to complete because it involves analyzing very large amounts of data and developing new methods. The impact of the proposed research is a deeper understanding of the relationship between differences in DNA sequence and variation in traits and diseases among individuals, as well as new methodological tools for analyzing massively large genetics data sets.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/dementia-risk-in-south-asians-data-from-the-uk

Dementia risk in South Asians: data from the UK

Last updated:
ID:
40055
Start date:
3 September 2018
Project status:
Closed
Principal investigator:
Dr Naaheed Mukadam
Lead institution:
University College London, Great Britain

Dementia causes a decline in brain functioning so that people become unable to do the things they previously could. It becomes more common as people become older, so as the world’s population ages, the number of people with dementia increases. The most common type of dementia is Alzheimer’s dementia and its risk is increased in older people who have a particular gene called APOE E4. There are other genes that also increase the risk of dementia but much less than APOE E4. The risk of dementia also increases with environmental risk factors, such as health conditions like diabetes and high blood pressure or with smoking. Although there has been a lot of research considering these environmental and genetic risk factors in people of European origin, there has been very little examining whether the environmental risk factors have the same effect in people from different ethnic backgrounds. They might not, as heart risk factors have different effects on the risk of stroke and heart disease in varying ethnicities and the gene APOE E4 may be less or more common in different populations.
I am interested in finding out whether people from different ethnic backgrounds have differing levels of dementia risk from environmental and genetic risk factors. I will focus on South Asian people, that is people from India, Pakistan and Bangladesh; comprising the largest non-White ethnic group in the UK.
I would use Biobank data to compare the frequency of APOE E4 and other known genetic variations relevant for dementia risk between White British and South Asian participants in UK Biobank participants. I will use it to compare the frequency of different risk factors, such as a diagnosis of diabetes and high blood pressure in South Asian people and White British people and how much each of those risk factors increases a person’s risk of dementia and whether this differs in different ethnic groups. Doing this research is important because very little is known about how dementia develops in minority ethnic groups. It may help to understand more about dementia in the general population and to find new ways of preventing or treating dementia.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/dementia-use-case-for-iasis-platform

Dementia use case for ‘iASiS’ platform

Last updated:
ID:
35916
Start date:
25 September 2018
Project status:
Current
Principal investigator:
Professor Peter Garrard
Lead institution:
City St George's, University of London, Great Britain

This work will contribute to the creation of a computational platform (iASiS) for integrating various data types (e.g. demographic, cognitive, imaging, genotyping) in dementia patients. The questions to be answered include: 1) how many patients who have been diagnosed with Alzheimer’s disease have (genetic) maternal/paternal family history and how many don’t? 2) Are there comorbidities that interact with the presumed pathology or the effects of family history and genetic determinants? 3) Are there any genetic markers of clinical variants (e.g. early onset; PCA; amnestic; MCI; psychiatric; language)? 4) Are there variant-specific prodromal syndromes (e.g. depression; anxiety)? The results of this study may lead to the identification of new biomarkers and (via the iASiS platform) interactions among known biomarkers, which will help improve the prevention, diagnosis and treatment of dementias and promotion of health throughout society.

The potential of identifying and integrating new biomarkers will also enrich the quality and quantity of information stored in UK Biobank and support future research. 1) Identification and clinical/radiological characterisation of dementia patients (Alzheimer’s Disease, Lewy Body Dementia, Frontotemporal Dementia) and age/gender matched controls with comorbidities, such as depression.
2) Stratification according to the type of dementia (primary) and the type of comorbidity (secondary).
3) When AD patients have coincident or premorbid depression, incidences of chromosome 3 p25-26 variant (linked to depression), and one or more APOE4 (linked to Alzheimer’s) alleles.
4) Presence of maternal/paternal history of dementia.
5) Comparisons of the proportions of maternal, paternal or no family history in each group. A subset of the full cohort consisting of people diagnosed with any form of dementia will be sufficient.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/demographic-characteristics-clinical-examination-biochemical-indicators-of-comorbidities-daily-routine-environmental-exposures-genetic-susceptibility-and-the-risk-of-musculoskeletal-disorders

Demographic characteristics, clinical examination, biochemical indicators of comorbidities, daily routine, environmental exposures, genetic susceptibility, and the risk of musculoskeletal disorders

Last updated:
ID:
154466
Start date:
9 June 2025
Project status:
Current
Principal investigator:
Dr Xinhua Qu
Lead institution:
Shanghai Jiao Tong University School of Medicine., China

Musculoskeletal disorders (MSDs), including diverse conditions affecting bones, joints, muscles, and connective tissues, pose a significant health challenge globally, impacting the lives of millions of individuals and carrying a substantial economic burden. Previous research has offered a range of interpretations regarding the underlying causes of MSD. We realize that not only demographic characteristics, living habits, environmental exposures and genetic characteristics can influence the occurrence of MSD, many comorbidities and chronic diseases of the heart, liver, intestines and brain also have an important impact on the susceptibility and development of MSD, which can be reflected by the biochemical indicators. Our study aimed to address these complexities and gaps by conducting a comprehensive, multi-dimensional, and long-term investigation into the trends of MSD. By examining these trends over the decades and exploring their connections with other relevant health conditions, our research provides valuable insights into the broader landscape of musculoskeletal health.

Our project is expected to last 36 months, and the causal evidence generated from the study will serve as a critical foundation for future research, healthcare strategies, and resource allocation, ultimately contributing to improved MSD prevention, management, and patient care. By staying proactive and responsive to these new challenges, healthcare systems can better serve the growing population affected by MSD.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/demographic-differences-for-blood-eosinophil-counts-and-the-clinical-importance-of-this-among-patients-with-asthma

Demographic differences for blood eosinophil counts, and the clinical importance of this among patients with asthma

Last updated:
ID:
667176
Start date:
24 February 2025
Project status:
Current
Principal investigator:
Miss Amy Shackleford
Lead institution:
Queen's University Belfast, Great Britain

Scientific Rationale
Over the last few decades, the increasing knowledge of the underlying inflammatory pathways in asthma has led to the development of monoclonal antibodies allowing greater disease control, minimizing OCS exposure and preventing accelerated lung function decline. High blood eosinophil counts are associated with more severe asthma exacerbations and poorer asthma control. Consequently, within the UK this biomarker is used as a key criteria for accessing biologic medications. The same threshold (typically >300 cells/uL) is currently applied to all patients, however evidence from the US that blood eosinophils differ by demographic factors such as ethnicity and gender has raised questions around whether this reflects inequality of access to biologic therapy. Further UK-focussed work is required to understand variations in blood eosinophils across the population, and their clinical implications among patients with asthma.

Research questions and objectives
The primary aim of this study is to investigate whether blood eosinophil counts differ between different demographic variables. Our secondary analysis will investigate whether demographically-adjusted blood eosinophil counts have higher predictive power when modelling asthma exacerbations than absolute blood eosinophil counts.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/demographic-lifestyle-and-genetic-factors-underlying-the-major-hyperlipidemias-and-the-associated-cardiovascular-risk

Demographic, Lifestyle and Genetic Factors Underlying the Major Hyperlipidemias and the Associated Cardiovascular Risk

Last updated:
ID:
62436
Start date:
16 March 2021
Project status:
Closed
Principal investigator:
Dr Iftikhar Kullo
Lead institution:
Mayo Clinic, United States of America

Lipid abnormalities including hypercholesterolemia, hypertriglyceridemia, and mixed hyperlipidemia are risk factors for heart attack and stroke. It is important to understand what genetic and lifestyle factors predispose to these lipid abnormalities and the associated cardiovascular risk. Previous studies have focused on genetic or lifestyle factors contributed to these abnormalities, but a comprehensive study addressing both in the same cohort is lacking. Additionally, these studies have been affected by several biases such as referral bias which means that only patients who were referred to specialty clinics were often studied. Such patients are not representative of the broader population. Using UK Biobank, we will be able to identify lifestyle and genetic factors related to these dyslipidemias, relatively free of referral bias. We will also quantify the risk of heart attack, stroke, and peripheral circulation disease due to these disorders.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/depression-and-aortic-stenosis-a-new-brain-heart-connection

Depression and aortic stenosis: a new brain-heart connection?

Last updated:
ID:
240506
Start date:
4 November 2024
Project status:
Current
Principal investigator:
Dr Giovanni Riccardo Maria Civieri
Lead institution:
University of Padua, Italy

The aortic valve divides the heart from the aorta. In Europe and North America, aortic stenosis is the most common primary valve lesion requiring surgery or transcatheter intervention. Its prevalence is rising rapidly because of the aging population, and approximately 10% of individuals aged > 80 years suffer from aortic stenosis. Currently, there is no medical therapy to prevent or treat aortic stenosis, and the only therapeutic option is to replace the native nave with a prosthesis, either surgically or percutaneously. Depression is highly prevalent among adults worldwide, and approximately 18% of individuals living in developed countries suffer from depression. Depression is an emerging risk factor for atherosclerotic cardiovascular disease. Although a clear mechanism underlying this association has not yet been demonstrated, depression could elicit different pathways that could ultimately lead to atherosclerosis, including endothelial and platelet dysfunction, altered autonomic nervous function, and increased systemic inflammation. Notably, inflammation is also highly involved in the pathogenesis of aortic stenosis and different systemic disorders (such as rheumatoid arthritis and psoriasis) are associated with higher incidence of aortic stenosis. Moreover, PET imaging allowed us to discover how inflammation of the valve predicts subsequent stenosis. We believe that depression also increases the risk of aortic stenosis by inducing systemic inflammation. To test our hypothesis, we will use data from the UK Biobank and assess whether depressed patients without aortic stenosis are at a higher risk of developing this valvular disease compared to similar patients without depression. Moreover, given that the association between depression and cardiovascular disease is stronger in women, we will also assess sex differences. If our hypotheses are confirmed, depression will be recognized as a new risk factor for aortic stenosis with important public health implications. First, clinicians will know that patients with depression are at a higher risk of developing aortic stenosis and will establish a proper follow-up. Second, new research lines to assess whether the treatment of depression reduces the risk of aortic stenosis will be implemented. Presumably, the project will last for three years. No controversial results are expected.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/derivation-of-a-machine-learning-ml-model-to-improve-pre-test-probability-of-identifying-individuals-with-genetic-variants-linked-to-familial-hypercholesterolemia-fh

Derivation of a machine learning (ML) model to improve pre-test probability of identifying individuals with genetic variants linked to familial hypercholesterolemia (FH).

Last updated:
ID:
67789
Start date:
27 January 2021
Project status:
Current
Principal investigator:
Mr Christophe Anne-Marie Thierry Stevens
Lead institution:
Imperial College London, Great Britain

Familial hypercholesterolaemia (“FH”) is an inherited disease caused by variations in genes related to the clearance of LDL-cholesterol (the so-called “bad cholesterol”). As a result, FH increases the levels of LDL-cholesterol from birth within the blood stream. Over time, this continuous exposure to high LDL-cholesterol results in fat deposits (“atherosclerotic plaques”) within the arteries, generally referred to as atherosclerosis. When these plaques become large and/or unstable, they can slow down the blood flow or generate blood clots obstructing the blood flow, causing heart disease or an acute heart attack. Cholesterol-lowering medications decrease levels of LDL-cholesterol, and, when administered early using effective doses, can prevent the development of atherosclerosis and heart diseases/attacks.

FH affects approximately 1 in every 311 individuals but is underdiagnosed, with less than 7% currently identified in the UK. Genetic testing is the most accurate tool for diagnosing FH, but it is expensive and not available everywhere due to a lack of resources. Diagnostic tools called clinical criteria are often used instead of genetic testing in daily clinical practice. These tools use patients’ characteristics including cholesterol levels, age at onset of heart diseases and family history, to make a diagnosis. Unfortunately, these tools might not work well in different populations and often fail to accurately identify FH patients.

With our research, we aim to help find FH patients using a branch of Artificial Intelligence called Machine Learning (ML). ML consists of a set of techniques that allow the replication a specific human behaviour involving reading large amounts of information and making predictions based on the data. ML models are computer software and mathematical models derived from the data that can differentiate between disease-free individuals and affected patients. We believe that ML models can better identify FH patients than clinical diagnostic tools currently used in clinical practices.

The performance of ML models will be compared to the performance of traditional clinical diagnostic tools. This will be done by counting the number of patients who have been misclassified by current diagnostic tools and newly derived ML models. If our ML models outperform clinical diagnostic tools, they will help identify more FH patients, on a national scale and earlier in life, ultimately allowing clinicians to treat more patients and help prevent heart disease. The present proposal over 3 years would be expected to substantially improve the current detection rates of <7% UK and <5% globally, in a cost-effective, scalable fashion.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/derivation-of-fitness-indicators-and-their-relation-with-hemodynamic-factors

Derivation of fitness indicators and their relation with hemodynamic factors

Last updated:
ID:
408
Start date:
1 December 2013
Project status:
Closed
Principal investigator:
Dr Soren Brage
Lead institution:
University of Cambridge, Great Britain

Few large-scale epidemiological studies have included measures of cardio-respiratory fitness, but those that have indicate that it is strongly related to metabolic disease, cardiovascular outcomes, some cancers, and all-cause mortality.
Some recent estimates from Health Survey for England (2008) indicate mean levels of fitness to be around 32 and 36 ml O2/min/kg for adult women and men, respectively. It is generally agreed, however, that this represents an overestimate due to stringent exclusion criteria (>40%).
The exercise test used in UK Biobank was designed to be as inclusive as possible to maximise the potential for linkage with incident disease. More specifically, exercise protocols were individualised to take into account participant characteristics including risk category.
This proposal aims to derive fitness indicators from information collected during the exercise test, and describe their variation by appropriate subgroups. Finally, we aim to demonstrate the etiological utility of derived fitness measures by examining the cross-sectional association with blood pressure and arterial stiffness.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deriving-biomarkers-for-menopause-the-search-for-modifiable-factors-to-delay-ovarian-aging

Deriving biomarkers for menopause: the search for modifiable factors to delay ovarian aging.

Last updated:
ID:
712057
Start date:
10 June 2025
Project status:
Current
Principal investigator:
Dr Frida Polli
Lead institution:
Massachusetts Institute of Technology, United States of America

The ovary is one of the first organs to age in the human body. At birth, ovaries contain all the eggs they will ever have, representing their “ovarian reserve”. The ovarian reserve dictates the reproductive lifespan as it undergoes a natural winnowing over time. Menopause is caused by the depletion of ovarian reserve (Johnson, Emerson and Lawley, 2022; Meng et al, 2018).
The ovaries are both pacemakers of healthy aging in women, as well as early warning systems of future pathologies. Despite great variability in age of natural menopause (ANM), and its importance as a marker for overall health, we are currently not able to predict its onset and our understanding of the correlates of ANM is rudimentary. The factors driving ovarian aging and therefore menopause remain poorly understood. We are interested in understanding the contribution of modifiable versus non-modifiable ones. Modifiable ones include hormonal, metabolic, reproductive and lifestyle factors while non-modifiable ones include genetics and gestational factors.
We will use machine learning to derive various algorithms that will serve as biomarkers of ANM. We will use ML best suited for regression analyses (eg random forest, support vector regression and k-nearest neighbors), prioritizing algorithms that allow for explainability of the model. We will build multiple ML models in stages: 1) blood makers only; 2) blood and questionnaire data on lifestyle factors; 3) blood, lifestyle and physiological data; 4) blood, lifestyle, physiological and genetic data.
We will use the following features:
! Blood markers: CBC (n=31); NMR measures (n=249); proteomics data (n~2,900)
! Reproductive factors: Gynecological and obstetric history
! Lifestyle factors: Smoking, alcohol, diet, exercise
! Physiological (VO2 max) and body composition scores
! Genetic Risk Score: A polygenic risk score (PRS) for ANM.
! Gestational factors: Singleton versus multiple birth; low birth weight.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deriving-novel-markers-of-general-and-hearing-health-from-measures-of-speech-perception-in-noise

Deriving novel markers of general and hearing health from measures of speech perception in noise

Last updated:
ID:
61759
Start date:
16 September 2020
Project status:
Closed
Principal investigator:
Dr Ian Michael Wiggins
Lead institution:
University of Nottingham, Great Britain

The UK Biobank holds information about hearing and cognitive ability from a large group of people, measured at different points in time. Hearing ability was measured using both a questionnaire and a listening test, the “Digit Triplet Test” (DTT), which measures the ability to understand speech in a background of noise. The UK Biobank also collected detailed data about the participant’s cognitive ability on the day of the listening test and how well they performed the test. There is currently little information about how much the results and performance on the listening test vary when it is performed repeatedly over time and whether it is reliable. The relationships between the changes in a person’s cognitive and hearing ability over time or their performance on the listening test are also not well understood but have the potential to help identifying new ways of measuring and managing hearing disability. Moreover, the use of large-scale data about hearing disability with computational models has the potential for identifying subpopulations that may require specific hearing treatments or to improve the predictions of developing problems with cardiovascular health that is important for good functioning of the hearing organ.

The present study will assess the variability and reliability of the measures of hearing ability in the UK Biobank. The detailed data about the listening test will be explored to determine potential new measures of hearing health and subpopulations of patients with specific profiles of hearing disability. The study will also explore the associations between measures of cognitive and hearing ability, and performance in listening tasks. The findings will be used to assess their utility for predicting the risk of health conditions associated with hearing health such cardiovascular disease.

The findings will help the researchers using listening tests and other hearing-related measures, including those in the UK Biobank, to better design and interpret the findings of their studies. The findings will also help to better understand the associations between hearing, cognitive and cardiovascular health, and to inform the development of new and personalised treatment strategies in the future.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/deriving-process-specific-polygenic-and-multi-omics-scores-of-cardiometabolic-risk-and-examining-their-association-with-medical-images

Deriving process-specific polygenic and multi-omics scores of cardiometabolic risk and examining their association with medical images

Last updated:
ID:
554976
Start date:
17 February 2025
Project status:
Current
Principal investigator:
Miss Hui Li
Lead institution:
Chalmers University of Technology, Sweden

Research questions:
1. How are process-specific polygenic scores (PPS) and multi-omics clusters associated with established cardiometabolic risk markers and preclinical tissue alterations?
2. Can deep regression and generative models based on multi-omics risk scores and MR imaging data reveal structural tissue changes associated with cardiometabolic disease progression?

Objectives:
Our first aim is to examine the association of process-specific molecular risk factors with established cardiometabolic risk markers. Our second aim is to explore the association of PPS and multi-omics clusters with MR imaging of cardiometabolic tissues.

Scientific Rationale:
Cardiometabolic diseases are multifactorial, involving both genetic and environmental risk factors. Recent studies have identified several cluster-speci!c partitioned polygenic scores (PPS) to decompose genetic risks into distinct underlying biological processes, offering insights for personalized prevention and treatment. However, genetics alone only explains part of cardiometabolic risk variability. Multi-omics data, such as proteomics, metabolomics, and microbiota, add critical insights by capturing the interaction between genetic predispositions and lifestyle factors during disease progression.
Advances in medical imaging, combined with deep learning techniques, enable detailed assessments of preclinical tissue anomalies, such as coronary artery calcification, plaques and ectopic lipid deposits. These advancements provide valuable insights for proactive prevention and management of cardiometabolic conditions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/description-of-cardiovascular-phenotype-in-the-uk-biobank-population-based-on-cardiovascular-magnetic-resonance-and-carotid-ultrasound

Description of cardiovascular phenotype in the UK Biobank population based on cardiovascular magnetic resonance and carotid ultrasound

Last updated:
ID:
2964
Start date:
1 September 2015
Project status:
Current
Principal investigator:
Professor Steffen Erhard Petersen
Lead institution:
Queen Mary University of London, Great Britain

Imaging of the heart and blood vessels is performed in a large subset of the UK Biobank cohort. Many measures defining the state of the heart and blood vessels can be derived from the images acquired. These measures are influenced by various health conditions and modifiable and non-modifiable factors, such as age, gender and ethnicity. The aim of this proposal is to describe the measures of the heart and blood vessel in the UK Biobank population and investigate how much modifiable and non-modifiable factors influence them. All new data will be made available for future research. Knowing the reference ranges for common imaging measures of the heart and circulation and how they are influenced by factors, such as age, gender, ethnicity, risk factors for heart attacks and strokes, is key for improving making diagnoses and predicting health outcomes. Descriptive statistics will be performed for all image derived phenotypes (IDPs) from the cardiovascular magnetic resonance (CMR) and carotid ultrasound images. We will perform subgroup analysis for important clinical factors, such as age, gender, cardiovascular risk, chronic conditions (e.g. Diabetes). We will apply descriptive statistics to a subpopulation considered `healthy without cardiovascular disease or presence of modifiable risk factors`. Univariate and multivariate regression analysis will be used to assess relationships between IDPs and relevant co-variates. We will also assess intra- and inter-observer variability for IDP measurement when repeat analysis is available. Initial 5000 subjects from the imaging enhancement study.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/description-of-multiple-risk-factors-for-end-organ-diseases-in-the-uk-biobank-population

Description of multiple risk factors for end-organ diseases in the UK Biobank population.

Last updated:
ID:
1002466
Start date:
5 November 2025
Project status:
Current
Principal investigator:
Dr Marcello Ricardo Paulista Markus
Lead institution:
University Medicine Greifswald, Germany

Although there were important improvements in treatment of cardiovascular diseases (CVDs) morbidity and mortality in the course of recent decades, the CVDs burden continues to be very significant. CVDs are multifactorial and linked to complex metabolic pathways. The identification of holistic risks and protective factors for CVDs continues to be a priority in epidemiological research. Previous studies considered associations of inflammation, physical fitness (determined by cardiorespiratory and muscle fitness) and cachexia were associated with increased mortality. Moreover, recent studies identified dietary patterns (such as plant-based and high-fat), beyond the isolated effect of single nutrients on lipid subfractions, and early life stressors, such as childhood maltreatment (CM), as significant contributors to long-term cardiometabolic risk, including atherosclerosis, hypertension, diabetes, hepatic steatosis, obesity, and renal dysfunction. The combination of CM and depression, particularly through sex-specific mechanisms, is still not well explored. Besides that, several organs dysfunctions such as various brain diseases, including cognitive disorders, dementia and Alzheimer’s, decreased renal function, and hepatic steatosis were associated with CVDs. Noteworthy, cardiovascular risk factors that have been linked to impaired function in one organ can thus result in alterations in the other and contribute to an acceleration of the ageing process in each organ. The aim of this proposal is to analyse, sex-specifically, how much modifiable and non-modifiable traditional risk factors, such as genomics, hypertension, T2D, hepatic steatosis, obesity, complete lipid profile, inflammation, physical fitness, and renal dysfunction, and new risk factors, such as cachexia, dietary patterns (plant-based and high-fat), and childhood maltreatment (CM), influence end-organ (brain, heart, kidney, liver) diseases and the interplay between these organs and their biological ageing.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/design-and-develop-novel-multi-omic-panel-for-preclinical-screening-and-early-diagnostics-for-neurodegenerative-disorders

Design and develop novel multi-omic panel for preclinical screening and early diagnostics for Neurodegenerative disorders

Last updated:
ID:
1078373
Start date:
4 November 2025
Project status:
Current
Principal investigator:
Dr Ashwini Rajasekaran
Lead institution:
BioOmicSignatures Tech Pvt. Ltd., India

In neurodegeneration, epigenetic modifications are emerging as stable, disease-relevant signatures that appear early in disease progression. cfDNA, released during cellular turnover and injury could act as a potential biomarker capable of detection in the preclinical phase. However, only about 2% of total cfDNA in circulation is neuron-derived. The low abundance makes detection difficult, contributing to why this area remains underexplored and yet to be integrated into clinical screening for neurodegeneration. Hence, we aim to develop a blood-based cfDNA methylation signature panel (identified using machine learning) using sensitive technology like nanopore. In addition, we want to improve this panel’s specificity and sensitivity by incorporating other omic markers into the panel for neurodegeneration.
Objectives:
* Identify neuron-specific cfDNA methylation markers, along with other omic signatures using machine learning analysis of large-scale public methylome datasets.
* Validate shortlisted markers with Oxford nanopore sequencing for high-resolution methylation profiling.
* Translate validated markers into a clinically implementable assay
Rationale:
The absence of reliable, minimally invasive diagnostics for neurodegeneration represents a critical barrier in clinical neuroscience. While imaging and CSF-based tests provide diagnostic information, their invasiveness, cost and late-stage applicability limit their impact. Protein markers are typically detectable only at later stages of the disease, limiting their utility for early intervention.
Omic signatures offers a transformative alternative, with the ability to detect neuron-derived signals from peripheral blood. Importantly, cfDNA methylation profiling would enable determination of cell-type origins, surpassing protein biomarkers in specificity. Established cfDNA-based assays in oncology, prenatal care, and transplantation, demonstrates their reliability as non-invasive diagnostics.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/design-of-new-indicators-for-co-morbidity-screening-based-on-stratified-risk-factors-for-cardiovascular-kidney-metabolic-syndrome-ckm-and-their-genome-sequence-data

Design of new indicators for co-morbidity screening based on stratified risk factors for cardiovascular-kidney-metabolic syndrome (CKM) and their genome sequence data.

Last updated:
ID:
866163
Start date:
4 July 2025
Project status:
Current
Principal investigator:
Mr Tang Qian
Lead institution:
Zhejiang University, China

Aims: Our research focuses on exploring the staging risk factors and genome sequence features that influence cardiovascular-renal-metabolic syndrome (CKM). Our goal is to screen high-risk populations for multidimensional risk factors, as well as to develop new screening metrics for different CKM staging to facilitate risk stratification and prognostication so that timely interventions and treatments can be implemented.

Scientific rationale:  Cardiovascular, renal and metabolic diseases have long been a major global public health challenge due to their high prevalence and mortality rates, as well as the enormous economic burden they impose on society. The American Heart Association (AHA) has proposed a new conceptual framework for chronic kidney disease (CKD) that aims to enhance the comprehensive and longitudinal management of CKD. Despite the growing understanding of these diseases, their etiology, pathogenesis and prognosis remain to be fully elucidated. The aim of this proposal is to analyze multidimensional risk factors associated with CKM using high quality data from the UK Biobank. The identification of risk factors, including genome sequence data, is expected to improve understanding of the pathophysiology of CKM and contribute to the development of more effective prevention and treatment strategies.

Project duration:  This project is expected to last for 36 months.

Public health impact:  This study aims to identify the risk factors associated with the development of these diseases and hopes to screen high-risk populations and obtain new screening indicators. It is expected that clinical decision guidance for treatment planning will help to reduce the economic burden on society and make a significant contribution to the field of public health.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/designing-a-resampling-approach-to-control-for-population-stratification-in-rare-variant-association-tests

Designing a resampling approach to control for population stratification in rare variant association tests

Last updated:
ID:
34716
Start date:
14 September 2018
Project status:
Closed
Principal investigator:
Professor Shengying Qin
Lead institution:
Shanghai Jiao Tong University, China

Recent technology advances have enabled large-scale analysis of rare variants, but properly testing rare variants associated with complex disorders remains a significant challenge. Most rare variant testing methods assume samples from a single population, which is often not true for large studies such as UK Biobank. We propose a population-informed resampling method for studies of multiple populations for rare variant tests. Preliminary results showed this empirical approach can effectively control for false-positives while maintaining statistical power. We plan to apply this method to various human complex disorders and traits including cancer, heart diseases, stroke, diabetes, arthritis, osteoporosis, eye disorders, autoimmune inflammatory, depression and forms of dementia. The findings will help us further elucidating biological mechanism of threatening illness, eventually lead to new insight into drug research and development. The project duration will be around 2 years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/designing-a-screening-program-for-nonalcoholic-fatty-liver-disease-in-the-primary-care-population

Designing a Screening Program for Nonalcoholic Fatty Liver Disease in the Primary Care Population

Last updated:
ID:
50689
Start date:
23 October 2019
Project status:
Closed
Principal investigator:
Dr Scott Andrew McHenry
Lead institution:
Washington University in St. Louis, United States of America

Nonalcoholic fatty liver disease is the liver-complication of obesity. It is the fastest growing cause of liver cirrhosis and liver cancer. It is also strongly associated with heart disease and diabetes mellitus, where a fatty liver often can be found years before those conditions are present. If a fatty liver can be reversed by diet, exercise or medications, it is likely that we can prevent these complications. Unfortunately, it is not as simple as imaging everybody since these tests (CAT scans, ultrasounds) are not very good at picking up subtle degrees of liver fat that still predispose to these conditions.

We have developed a risk-score based on routinely obtained blood work for nonalcoholic fatty liver disease to identify patients at an early stage before they develop complications such as diabetes mellitus or liver cirrhosis. Before this can be used in primary care clinics, we must confirm that it is predictive in more than one country, which is why we wish to validate the score in the UK biobank.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/designing-and-validating-cancer-risk-and-prognostic-tools

Designing and validating cancer risk and prognostic tools

Last updated:
ID:
61181
Start date:
7 February 2022
Project status:
Current
Principal investigator:
Dr Julia Steinberg
Lead institution:
Cancer Council NSW, Australia

Cancer is a disease with huge health burden. In the UK alone, every two minutes someone is diagnosed with cancer, and the numbers have been rising over the past decade. Therefore, it is important to identify factors that contribute to cancer risk. In this project, we will conduct analyses to identify genetic, lifestyle, environmental, demographic and health factors that are associated with cancer risk. To enable targeted and personalised prevention approaches, we will investigate how the identified risk factors can be used to stratify individuals for risk-appropriate screening – for example, whether a particular risk score for breast cancer can accurately identify women who would benefit from more frequent or earlier screening. As it is known that the prediction of risk using genetic data can be less accurate for people with non-European ancestry, we will quantify differences in the quality of cancer risk predictions for people of different ancestries. Where differences are identified, we will examine how the risk prediction can be improved for people of diverse ancestries.

Where feasible, we will compare the insights gained from this work with comparable analyses of other international data, such as the 45 and Up Study cohort in Australia, which includes over 250,000 residents of New South Wales, aged 45 and over. This will allow us to determine the extent to which factors influencing cancer risk are similar or different across countries and settings. The comparison will also allow for risk tools developed based on Australian data to be independently tested using the UK Biobank data, and vice versa.

In a second strand of this work, we will follow a similar approach to investigate factors associated with death after a cancer diagnosis. In particular, we will examine associations between COVID-19 and cancer, and changes in survival of people with cancer due to the COVID-19 pandemic.

This study will improve the understanding of factors contributing to cancer risk and prognosis, paving the way for improved interventions to reduce the future burden of cancer. By studying potential differences in the quality of risk prediction between ancestry groups, this work will also highlight to what extent the use of such new risk tools could exacerbate existing health inequities. Thus, this work is very well-aligned with the aim of UK Biobank to improve prevention, diagnosis and treatment of a wide range of serious and life-threatening illnesses.

The intended duration of this project is 5 years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/designing-optimal-neuromodulation-strategies-for-depression-using-neuromorphic-computing-inspired-by-multimodal-brain-imaging-data

Designing optimal neuromodulation strategies for depression using neuromorphic computing inspired by multimodal brain imaging data

Last updated:
ID:
447069
Start date:
8 May 2025
Project status:
Current
Principal investigator:
Professor Yuxiao Yang
Lead institution:
Nanhu Brain-computer Interface Institute, China

This project aims to explore effective targets and stimulation paradigms for Deep Brain Stimulation (DBS) in the treatment of Major Depressive Disorder (MDD) through brain imaging and neuromorphic computing methods. We will address the current challenges in DBS clinical treatment for MDD, such as target brain region selection, biomarker identification, and stimulation parameter optimization, by focusing on the following aspects:
1. Brain Network Construction and Analysis:
* Based on brain imaging data, construct structural and functional brain networks using brain parcellation maps to study the relationship between individual differences in depression and changes in brain networks.
2. Study of Brain Iron Deposition and Depression:
* Using brain imaging data, explore the relationship between depression and brain iron deposition through brain susceptibility information, aiming to identify biomarkers of the pathophysiological mechanisms of depression.
3.Neuromorphic Computing and Simulation Stimulation:
* Utilize multimodal brain connectivity patterns and neurodynamic theoretical methods to establish a biologically constrained brain-inspired intelligence model. Simulate neural signals of depression patients and perform virtual patient stimulation to observe changes in brain functional networks, thereby exploring optimal targets and stimulation paradigms for DBS.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/detecting-anemia-using-fundal-retinal-images

Detecting anemia using fundal (retinal) images.

Last updated:
ID:
142339
Start date:
15 November 2023
Project status:
Current
Principal investigator:
Dr Vinay Arora
Lead institution:
Thapar Institute of Engineering & Technology, India

Our study seeks to provide a novel, non-invasive technique for diagnosing anemia, a common medical illness marked by a deficiency in red blood cells or low hemoglobin levels in the blood. Early identification is essential for efficient treatment of anemia because it can cause symptoms like weakness, exhaustion, and difficulties concentrating.

Our project’s scientific justification is that anemia can alter the small blood vessels in the retina of the eye, which can be photographed using retinal imaging. With the aid of cutting-edge computer techniques, we analyze these retinal images with the goal of developing a tool that can swiftly and precisely identify anemia.

The length of the project is anticipated to take approx. 03 years. The results of our study have a considerable effect on public health. If it works, this approach could provide a straightforward, affordable, and widely available screening tool, revolutionizing the identification of anemia. This suggests that more people might undergo anemia testing, particularly in regions with poor access to medical facilities. Anemia can be better managed with early discovery, possibly halting its progression and associated health problems. Finally, by making advanced diagnostics accessible to a larger public, our research has the potential to enhance the general health and wellbeing of people and communities, decrease healthcare costs, and advance health equity.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/detecting-causal-inference-associations-between-lifestyle-environmental-factors-genetics-and-noncommunicable-diseases

Detecting Causal Inference Associations Between Lifestyle, Environmental Factors, Genetics, and Noncommunicable Diseases.

Last updated:
ID:
314402
Start date:
11 November 2024
Project status:
Current
Principal investigator:
Professor Yingyi Qin
Lead institution:
Naval Medical University, China

The onset and progression of noncommunicable diseases (such as cardiovascular diseases, renal diseases, metabolic related diseases, cancer, etc.) is associated with lifestyle, environmental factors, and genetic factors. The main purpose of the study is to clarify the causal effects of these factors in the onset and progression of diseases, providing relevant data-based evidence for disease prevention and health guidance by using causal inference analyses methods. We will apply propensity score methods (matching/ weighting/ doubly robust model/ propensity score method with machine learning algorithms), instrumental variable model, causal mediation analysis methods to indicate the pathway of causal inference between factors and outcomes (onset or progression of noncommunicable diseases). Meanwhile, we will also verify the applicability of the causal inference models we have constructed.
Additionally, we will attempt to construct prediction model by using machine learning methods, as well as transfer learning or deep learning methods based on the identified factors.
Our study will span 36 months, and we plan to utilize the whole cohort dataset.
This study will provide accurate data-based evidence/suggestion for disease prevention and improving people’s status/level, and we also establish precise prediction models to support disease forecasting.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/detecting-early-imaging-phenotypes-of-tau-associated-dementia-in-the-general-population

Detecting early imaging phenotypes of tau associated dementia in the general population

Last updated:
ID:
46620
Start date:
5 September 2019
Project status:
Closed
Principal investigator:
Dr Timothy Rittman
Lead institution:
University of Cambridge, Great Britain

Progressive Supranuclear Palsy (PSP) and Frontotemporal Dementia (FTD) are diseases that cause a devastating impact on their sufferers, resulting in progressive cognitive decline, problems with walking and other movements, and significantly reduced life expectancy. Currently no intervention exists that can stop or slow down the advance of disease in these conditions. The multiple failures in trials of treatment of various forms of dementia have in part been due to difficulties in recognising and targeting treatment at individuals with early or presymptomatic disease.

Modern imaging techniques have demonstrated that brain network connections and brain structure are altered in patients with PSP and FTD. However, the order in which these changes appear over time is unclear, as is the extent to which they precede the onset of symptoms.

We have characterised the structural imaging, functional imaging and cognitive changes seen in these conditions from a large cohort of patients with PSP and FTD seen in Cambridge. Our aim is to apply these imaging and cognitive testing derived “fingerprints” to identify a section of the general healthy population who most closely resemble those with the disease using a machine learning approach. We will then use outcome data from UK biobank to further characterise this group and their comparative health outcomes. This will give us an insight into how these markers change early in disease and in the general population, and the order in which any such change occurs.

The potential to identify an at-risk population can inform novel trials of therapeutic intervention together with facilitating diagnosis, prognosis and monitoring of treatment.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/detecting-gene-gene-interaction-effect-on-blood-cell-traits-by-neural-networks

Detecting gene-gene interaction effect on blood cell traits by neural networks

Last updated:
ID:
46791
Start date:
9 May 2019
Project status:
Closed
Principal investigator:
Dr Pekka Marttinen
Lead institution:
Aalto University, Finland

The main purpose of Genome-Wide Association Studies is to understand the relationship between human genes and diseases. One approach is to analyze the relationship between human genes and metabolites or cell traits, which are highly related to most diseases. The effect of genes on traits can be divided into two parts: the main part (effect of each gene alone), and the interaction part (interaction effect between different genes). When the number of relevant genes is large (very common for most traits), the number of possible interactions grows rapidly, and detecting all possible interaction pairs can be intractable for traditional methods.

This study focuses on developing modern neural networks algorithms which can detect gene-gene interactions on blood cell traits efficiently. Neural network algorithms have been successfully used in many high-dimension datasets such as images, speeches, and languages, and detecting statistical interactions by neural networks have also been widely studied in recent years. But in statistical genetics, few approaches have taken the advantage of ‘Big Data’ by using deep neural networks.

This project will take about 1 year to develop related algorithms and test on blood cell traits data. Our work can also be generalized to study interactions on metabolites data, leading to interpretable relations between genes and diseases. The tool developed in this work has the potential to analyze other kinds of interactions, such as gene-environment interactions and gene-drug interactions, which can be a foundation for developing new drugs for many diseases in a genetic level.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/detecting-genetic-effects-on-the-variability-of-cardiometabolic-risk-profiles-and-clinical-outcomes

Detecting genetic effects on the variability of cardiometabolic risk profiles and clinical outcomes

Last updated:
ID:
112124
Start date:
30 October 2023
Project status:
Current
Principal investigator:
Dr Jordana Bell
Lead institution:
King's College London, Great Britain

Aims: This project aims to detect the genetic effects on the variability of cardiometabolic risk profiles and related clinical outcomes. The project will also explore whether these signals can capture gene-gene (GxG) and gene-environment (GxE) interactions underlying cardiometabolic disease risk.

Scientific rationale: Obesity is a major public health concern and a key risk factor for cardiometabolic disease. Obesity is influenced by multiple genetic variants, environmental factors, and their interactions. Previous studies have identified evidence for both GxG and GxE interactions underlying obesity. Identifying such interactions is challenging, but the results better characterise the genetic architecture of obesity and its associated disease risk. Furthermore, the presence of GxE interactions suggests potential for targeted lifestyle interventions in individuals of particular genotypes to reduce obesity levels and susceptibility to cardiometabolic disease.

The presence of GxG or GxE interactions leads to an increase of phenotypic variability. Therefore, detecting the genetic effects on trait variability (vQTLs) is an alternative approach to capture GxG and GxE interactions. Previous vQTL studies of cardiometabolic traits identified several GxE interactions in UK Biobank (Wang et al. 2019 Science Advances, Westerman et al. 2022 Nature Communication, Lu et al. 2022 Genetics). These studies focused on anthropometric measures, such as BMI, and circulating cardiometabolic risk biomarkers. Here, we expand this work by considering additional measures of adiposity including body fat distribution measured by MRI and DXA. This is of particular relevance to cardiometabolic health because central adiposity has stronger association with metabolic disease than whole-body fat. Overall, this project seeks to detect vQTLs for multiple measures of adiposity, body fat distribution, cardiometabolic risk biomarkers, and related clinical outcomes. If vQTLs are detected, follow up analyses will explore evidence for GxG and GxE interactions at selected signals. GxG analyses will consider interactions with previously detected GWAS signals. GxE interaction analysis will be conducted with multiple environmental factors, including smoking, exercise, dietary intakes, and others. Lastly, the project will explore whether the vQTLs, and GxG and GxE interaction effects can be incorporated in polygenic risk scores to improve the prediction of cardiometabolic traits and disease.

Project Duration: The project forms part of a PhD thesis and should be completed within 3 years.

Public Health Impact: Our findings could provide novel insights into gene-environment interactions of cardiometabolic risk and clinical outcomes. The current study may also help to develop personalized prevention and prediction strategies for obesity and cardiometabolic disease risk.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/detecting-heterogeneity-in-gwas-traits-from-expected-correlations-between-polygenic-risk-score-predictors

Detecting heterogeneity in GWAS traits from expected correlations between polygenic risk score predictors

Last updated:
ID:
55739
Start date:
29 March 2020
Project status:
Closed
Principal investigator:
Professor Itsik Pe'er
Lead institution:
Columbia University, United States of America

Genome-wide association studies have in the past decade discovered many variants in the genome which inform an individual’s risk of various diseases. But conventional GWAS often labels individuals as strictly cases or controls for a disease. In practice, this may too reductive, as within a particular disease there may exist a distinct number of sub-types each with their own sets of observed symptoms. Some examples include bipolar disorder (manic or hypomanic), depression (typical and atypical), lung cancer, and breast cancer. Our goal is to detect the presence of these sub-types, which we term heterogeneity, without necessarily determining which individuals belong to which sub-type. By reducing the complexity of our model in this way, we have a decent chance with current cohort sizes to obtain the required signals, which are very small and widely dispersed throughout the genome. Our model CLiP (Correlated Liability Predictors) detects heterogeneity by calculating how often particular genetic variants co-occur with one another among affected individuals and compares these values to what we would expect when no heterogeneity is present. We expect the duration of this project to be 1 year. We have already devoted significant time to developing and validating CLiP with simulated GWAS cohorts, so this time will consist of data processing. While interest in disease heterogeneity has increased in recent years, there has been relatively little work on discovering hidden sub-types. We hope that CLiP is a first step toward routinely considering the possibility of heterogeneity when conducting GWAS. Additionally, recent works have shown that genetic variants associated with diseases may vary among different populations separated by sex or ancestry, and the overrepresentation of certain groups in GWAS may lead to disparities in diagnosis and care. It stands to reason that there may be numerous diseases with distinct genetic associations across different groups, many of which may not neatly divide among readily visible attributes. Addressing heterogeneity in this scenario will ensure we are properly serving every population.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/detecting-high-risk-smokers-in-primary-care-electronic-health-records

Detecting high-risk smokers in Primary Care Electronic Health Records

Last updated:
ID:
74471
Start date:
24 February 2022
Project status:
Current
Principal investigator:
Miss Lamorna Brown
Lead institution:
University of St Andrews, Great Britain

The aims of the project are to:
– Evaluate the quality and validity of EHRs in Scotland to identify and characterise patients with lung cancer.
– Develop a risk model using information from EHRs.

With the main research question:
What factors/data features, contained in electronic health records (EHRs), are associated with and produce estimates of risk for lung cancer in individuals that smoke?

Rationale and impact
Although lung cancer screening has been recommended to aid in the early detection of lung cancer, it is yet to be implemented. To enable implementation, further research exploring methods to identify eligible screening participants and optimum risk thresholds for inclusion would be required. As this project will examine whether Electronic Health Record (EHR) data can be used for the identification of smokers at high risk of developing lung cancer, it will address the need for further research.
EHRs can provide researchers with data on metrics and features that may be challenging to obtain using other methods. There is also compelling evidence that EHRs can provide researchers with information on smoking behaviour in individuals. The accuracy and validity of this data is yet to be identified, as this will vary depending on the reference used and the accuracy with which data is extracted. Regardless, use of EHRs in epidemiological research has the potential to reduce late lung cancer diagnosis and explore data features such as symptoms and pack years that would have otherwise been difficult to accurately obtain. As such, this project will aim to expand knowledge around lung cancer risk research by seeking to identify smokers in EHRs and subsequently utilising this information in modelling risk of incidence. If EHR information can be used to accurately predict risk of lung cancer, a risk prediction score can be developed which GPs can feasibly use to aid them in their identification of at risk patients.

Duration
The project will last 3 years.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/detecting-individuals-who-are-at-risk-of-developing-lifestyle-diseases

Detecting individuals who are at risk of developing lifestyle diseases.

Last updated:
ID:
102535
Start date:
9 June 2023
Project status:
Current
Principal investigator:
Mr Stefan Wijtsma
Lead institution:
Labonovum B.V., Netherlands

Our project at Labonovum B.V. applies machine learning in preventive health care in order to identify and notify individuals at risk of developing some lifestyle disease like diabetes type 2, Cardiovascular disease, obesity, Chronic obstructive pulmonary disease, osteoporosis, and asthma. We are also interested in detecting asymptomatic individuals who already have one of the previously mentioned diseases.
We believe that it is of great importance to detect those individuals at early stages of illness the earliest possible.
If they are at risk, our goal is to prevent the incidence of the disease later in life. If they are asymptomatic, our goal is informing them so they start treatment before their illness reaches a more severe stage.
Our motives are to reduce workload on doctors, reduce expensive healthcare costs, and increase the longevity and quality of life of individuals. Doctors in the Netherlands are facing a huge workload as there are not enough general practitioners. We contribute to decreasing that workload by reducing the number of lifestyle diseases patients, especially that those diseases are very much widespread. By preventing the incidence of those diseases, we reduce their treatment costs which are high. One of our motives is to impact public health in a helpful manner. Lifestyle diseases negatively impact the quality of life of patients as they are forced to go to the emergency room or visit doctors more often. We save our patients from suffering a poor lifestyle or even from death. The WHO (World Health Organization) estimates that 80% of premature deaths from cardiovascular disease and diabetes could be prevented by controlling of their risk factors. We conclude that our project increases longevity and quality of life of the public.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/detecting-pleiotropic-effects-through-integration-of-omics-data

Detecting pleiotropic effects through integration of omics data

Last updated:
ID:
32285
Start date:
10 October 2018
Project status:
Current
Principal investigator:
Dr Andrew DeWan
Lead institution:
Yale University, United States of America

Using omics data, we will attempt to identify shared genetic variants that play a role in a number of common traits and diseases that have a high public health significance that include: asthma, obesity, type 2 diabetes, blood pressure and blood lipid profiles. We will accomplish these goals by using the UK Biobank data and a second dataset with gene expression and genome sequence data on a small set of subjects. We will use statistical methods to detect shared genetic effects and perform biological validation in order to bring about a better understanding of the role shared genetics plays in complex disease. The co-occurrence of common traits and diseases pose a critical public health challenge; this co-occurrence may be explained, in part, by genetic loci shared between these traits and diseases.

Identifying shared genetic loci for pairs of traits/diseases will advance our understanding of the mechanistic links between them. These loci have the potential to serve as targets for a single intervention that simultaneously treats both diseases. We will implement previously developed statistical methods and extend existing methods to analyze imputed and rare genetic variants to identify variants associated with two of the diseases/traits listed in 1a. All methods will be implemented in software developed by one of the investigators on this proposal, Dr. Suzanne Leal, which uses parallel processing to make it feasible to analyze hundreds of thousands of samples efficiently and quickly. We plan to analyze the full cohort of approximately 500,000 subjects with genotype and phenotype data.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/detecting-rare-haplotype-association-with-multiple-correlated-continuous-phenotypes

Detecting Rare Haplotype Association with Multiple Correlated Continuous Phenotypes.

Last updated:
ID:
96020
Start date:
2 March 2023
Project status:
Current
Principal investigator:
Mr Ibrahim Hossain Sajal
Lead institution:
University of Texas (UT Dallas), United States of America

Health-related studies collect information on many traits/outcomes which are later used in the development and evaluation of treatment. Outcomes can be the disease status of an individual, or traits that are used as indicators of disease status. Systolic and diastolic blood pressures are examples of traits that are used to define hypertension. Several diseases/traits/outcomes are often correlated as these are collected on the same individual. Outcomes may also share a common underlying genetic mechanism. Hence, it is important to consider those outcomes jointly when trying to uncover the genetic variants associated with the outcomes. In particular, we are interested in finding the genetic variants that are associated with multiple brain imaging outcomes. We propose to develop new statistical methods to this end and apply them to the UK Biobank data.
Since the project is still in its infancy and we are only developing the method now, we need around 36 months to complete all the steps i.e. testing our method using simulations, comparing it to the existing association tests, application of the method to the UK Biobank data, and finally disseminating the findings of our project.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/detecting-sexually-antagonistic-conflict-across-the-human-genome

Detecting sexually antagonistic conflict across the human genome

Last updated:
ID:
75040
Start date:
24 May 2023
Project status:
Current
Principal investigator:
Professor Graham Coop
Lead institution:
University of California, Davis, United States of America

Humans have 23 pairs of chromosomes – 22 pairs of autosomes, and one pair of sex chromosomes. Male individuals usually have an X and Y chromosome, while females usually have two X chromosomes. Unlike the autosomes, the X and Y chromosomes spend different amounts of time in males and females across generations. Because the sex chromosomes spend different amounts of time in males and females, we expect them to display unique patterns of sexually antagonistic variation. Sexually antagonistic variation simply describes genetic variants that are either beneficial in females and costly in males, or beneficial in males and costly in females. As males and females have different physiological and morphological requirements, sexually antagonistic variation is expected to be very common throughout the genome, and, because it reflects selection acting in opposite directions in each sex, we predict that sexually antagonistic variation will evolve differently in regions of the genome that are not shared equally between the sexes. This project will investigate the distinct role of the sex chromosomes and other sex-biased genetic elements, such as the mitochondria, in promoting and maintaining sexually antagonistic variation. By studying regions of the genome that spend different amounts of time in males and females – those exceptions to the symmetric transmission of the autosomes – we can gain insight into how sexually antagonistic variation is maintained across the genome as a whole.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/detecting-unrecognised-dementia-across-ethnic-populations-development-and-validation-of-a-dementia-risk-prediction-model

Detecting unrecognised dementia across ethnic populations: development and validation of a dementia risk prediction model

Last updated:
ID:
739774
Start date:
19 August 2025
Project status:
Current
Principal investigator:
Ms Xiaochen Yang
Lead institution:
Chinese University of Hong Kong, China

As a global public health crisis, dementia imposes a substantial burden on individuals, healthcare systems, and societies worldwide. There are also large amounts of undiagnosed cases, particularly among ethnic minorities and low-income groups with limited healthcare access. All subtypes of dementia are progressive, and the duration from dementia diagnosis to death is short, underscoring the need for early identification and intervention. Current dementia prediction models suffer from limited generalisability due to reliance on invasive tests and underrepresentation of non-Western populations.
Research questions of this study: 1. What biological, sociodemographic and modifiable lifestyle factors synergistically contribute to dementia risk? 2. How do dementia risk profiles differ between the UK Biobank and East Asian cohorts?
The objectives of this study include: 1. To develop and validate a dementia risk prediction model for the UK Biobank cohort that integrates genetic factors, biomarkers, sociodemographic variables, and modifiable lifestyle factors. 2. To identify ethnic and cultural disparities in risk factor effect sizes by contrasting UK Biobank data with East Asian cohorts.
By integrating biomarkers with modifiable risk factors, this study could enhance the predictive power of dementia risk models and their real-world applicability. Identifying ethnic and cultural differences in risk factor prevalence and effect sizes will contribute to the development of tailored strategies for dementia prevention and early intervention.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/detection-and-risk-prediction-of-neuropsychiatric-disorders

Detection and Risk Prediction of Neuropsychiatric Disorders.

Last updated:
ID:
689286
Start date:
1 April 2025
Project status:
Current
Principal investigator:
Mr Yanheng Li
Lead institution:
Beijing Institute of Technology, China

By 2050, the global prevalence of Neuropsychiatric Disorders (NPDs), including conditions such as Alzheimer’s disease, schizophrenia, and other cognitive and affective disorders, is projected to reach approximately 4.9 billion cases, representing a 22% increase from 2021 estimates. This significant rise is attributed to factors such as an aging global population and increasing environmental stressors. These disorders pose substantial challenges for early diagnosis and intervention, as they often exhibit overlapping symptoms and complex neurobiological mechanisms.
Despite advancements in neuroscience and medical imaging, current diagnostic approaches remain limited in accuracy and accessibility, relying heavily on subjective clinical assessments and single modality biomarkers.
To address this gap, the present study aims to adopt an integrated approach by leveraging the rich radiomics, genetics, metabolomics, proteomics, and comprehensive epidemiological data available in the UK Biobank. Through multidimensional analyses, we seek to identify novel risk factors, determine potential biomarkers, develop single-modality or multimodal diagnostic models, and elucidate the causal relationships underlying various neuropsychiatric disorders. This comprehensive approach will facilitate more personalized and timely interventions, ultimately improving patient outcomes and advancing the field of neuropsychiatric diagnostics.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/detection-of-large-scale-copy-number-and-copy-neutral-mosaic-alterations-in-circulating-leukocyte-dna-of-uk-biobank-cancer-cases-and-cancer-free-controls

Detection of large-scale copy-number and copy-neutral mosaic alterations in circulating leukocyte DNA of UK Biobank cancer cases and cancer-free controls

Last updated:
ID:
21552
Start date:
1 August 2016
Project status:
Current
Principal investigator:
Dr Stephen Chanock
Lead institution:
National Cancer Institute, United States of America

We aim to investigate potential differences in frequency and location of large acquired copy number mutations in blood derived DNA of cancer cases and healthy controls. Our past research has demonstrated that the frequency of large, mosaic mutations increases with age, but has produced limited evidence for cancer associations. The UK Biobank is a well-powered collection of samples with the opportunity to provide new insights into how large acquired copy number mutations in the blood may be related to future cancer risk as well as perform a genome-wide association study of mosaic copy number alterations. Our proposed research compliments UK Biobank’s aim of improving the prediction and detection of serious and life-threatening illnesses such as cancer by investigating how an individual’s acquired copy number mutations may relate to their future risk of cancer. Our calling algorithms will scan the chromosomes of all UK Biobank participants with genotyping data to detect large, acquired mosaic events. Frequencies and locations of events will be compared across individual characteristics in univariate and multivariate models to better understand how acquired copy-number and copy-neutral events relate to cancer risk. We propose to use all participants from the full cohort with available SNP array genotyping data.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/detection-of-pleiotropic-effects-among-pharmacogenes-in-uk-biobank-participants

Detection of pleiotropic effects among pharmacogenes in UK Biobank participants

Last updated:
ID:
15357
Start date:
1 October 2015
Project status:
Closed
Principal investigator:
Professor Julie Hussin
Lead institution:
Montreal Heart Institute, Canada

Progress in the field of pharmacogenetics is increasingly allowing a better understanding of the molecular mechanisms modulating drug sensitivity, efficacy and toxicity. Preliminary evidence suggests that genetic variants in genes involved in these processes, known as pharmacogenes, can also lead to an altered risk of disease in carriers. Therefore, their diverse established and intertwined roles in metabolic pathways make them an ideal system to study pleiotropy, a phenomenon in which a single locus affects distinct traits. Here, we propose to use the UK Biobank data to systematically identify pleiotropic relationships among known pharmacogenes, taking advantage of innovative computational methods. The field of pharmacogenomics has become one of today’s most promising aspects of personalised genomics. Identification of variants that may have systemic disease risk as well as altered drug response would provide insights to help optimizing genetic testing strategies and drug therapy, leading to better prevention of adverse effects of treatment. Furthermore, identifying relationships between phenotypes and pharmacogenes, most of which have known functions, will lead to improved understanding of the molecular mechanisms of disease. We will select a collection of genes previously identified in pharmacogenomics studies that consists of established pharmacogenes, as well as GWAS hits for drug response. Phenotype data and electronic medical records information from participants will be used to perform statistical tests of association with each pharmacogenetic marker. As a first approach, the Phenome-Wide Association Study (PheWAS) methodology is appropriate to explore genotype-to-phenotypes associations. As this method harbours non-negligible methodological challenges, especially in properly defining the ?phenome? and in dealing with covariates, novel approaches to identify pleiotropic interactions will be developed and applied to this extensive list of pharmacogenes. Full cohort


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/determinants-and-clinical-significance-of-cerebral-small-vessel-disease

Determinants and clinical significance of cerebral small vessel disease

Last updated:
ID:
94113
Start date:
29 September 2023
Project status:
Current
Principal investigator:
Professor Stephanie Debette
Lead institution:
University of Bordeaux, France

Stroke and dementia are the most common age-related causes of death and disability, representing a huge burden for society. Cerebral small vessel disease (cSVD) is a group of diseases affecting the small vessels that supply blood to the brain. The brain abnormalities that characterize cSVD, and that are due to damage to these small vessels, can be seen on brain images, particularly Magnetic Resonance Imaging (MRI). With increasing age, cSVD becomes very common and is often “covert” in older adults, meaning that abnormalities are detectable on brain MRI but not associated with clinical stroke. Knowing that covert cSVD is a strong predictor of future stroke, cognitive decline and dementia in older adults, better detection and management of covert cSVD could have a major impact on preventing disability and costs associated with these neurological diseases. The current incomplete understanding of the biological mechanisms leading to cSVD is one of the reasons for the current lack of clinically useful “biological markers” and of specific treatments for this disease. Therefore, efforts to improve our understanding of the biological mechanisms causing cSVD and to identify both early disease markers and new drug targets for cSVD are of great importance.
We aim to use the UK Biobank data to: (1) develop imaging methods allowing to accurately quantify cSVD imaging abnormalities on brain MRI; (2) explore cardiovascular and retinal features of cSVD using cardiovascular and retinal imaging (leveraging the similarities between retinal and brain small vessels), and identify shared biological pathways; (3) discover novel genetic risk factors for cSVD as well as circulating blood molecular biomarkers such as proteins or metabolites; (4) improve risk prediction of stroke and dementia in persons with covert cSVD, based on imaging and circulating biomarkers, using novel statistical methods.
In terms of public health impact, this project will facilitate early diagnosis of cSVD to improve detection of at-risk subjects and prevent the occurrence of stroke and dementia. It will provide a better understanding of disease mechanisms and accelerate drug target discovery. In the long run, our research will contribute to the development of personalized preventive and therapeutic strategies for cSVD and its complications.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/determinants-and-consequences-of-brain-connectivity-gradient-variation-role-in-cognition-mood-mental-wellbeing

Determinants and Consequences of Brain Connectivity Gradient Variation: Role in Cognition, Mood & Mental Wellbeing

Last updated:
ID:
936468
Start date:
31 October 2025
Project status:
Current
Principal investigator:
Professor Dara M Cannon
Lead institution:
University of Galway, Ireland

Improving societal protection of brain health will involve substantially greater understanding of the factors that influence and are influenced by intricate patterns of brain connectivity across functional networks and cortical gradients. The present proposal represents a modest attempt to begin to define the role altered structural and functional brain connectivity relationships may have on human mood among other functions critical to human wellbeing such as cognition. Determinants such as genetics, personal (trauma) and community-based (deprivation) factors are likely to contribute to optimal patterns of brain connectivity and in turn, these may mediate outcomes critical to personal and social wellbeing. While many studies relate life adversity to mental wellbeing outcomes, the recent advances including our own defining connectivity gradients, offers the potential to investigate their role in mediating such relationships. Evidence derived from this proposal would support the development of effective policy interventions aimed at mental health and wellbeing in society and fundamentally advance our understanding of brain connectivity in relation to human functioning. Specifically, we will establish the contribution of genetic variation, personal and social factors such as trauma and deprivation to brain connectivity gradients of coupling (objective 1), investigate the relationship of connectivity patterns in mood and cognition (objective 2) and finally, construct an overall model including the determinants and consequences to examine the mediating role of brain connectivity gradients in mental wellbeing, ultimately aiming to identify modifiable factors that may support the development of brain health related research and policy development.

This research does not involve development of any proprietary AI model and will not in any way make participate level data available. TRIPOD AI guidelines apply to this research and will be adhered to in all analyses.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/determinants-and-environmental-correlates-of-asthma-attacks-in-uk-biobank

Determinants and environmental correlates of asthma attacks in UK Biobank

Last updated:
ID:
54874
Start date:
29 April 2020
Project status:
Current
Principal investigator:
Dr Elaine Fuertes
Lead institution:
Imperial College London, Great Britain

Asthma is the most common chronic respiratory disease worldwide. How often asthma attacks happen, as well as how severe they are, is influenced by a person’s sex, age, if they smoke, whether they have other allergic diseases such as hayfever and eczema, as well as many other factors. Some people suffer asthma attacks when they breathe in allergens (small particles capable of causing allergic reactions, such as pollens and moulds), and when traffic pollution is high. If exposed to both, their asthma may become very severe as breathing in traffic pollution can make the lungs especially sensitive. Further, plants growing in high traffic areas appear to release more allergen than plants growing away from traffic.

This 3-year project will identify determinants that influence the frequency and severity of asthma attacks and the occurrence of other allergic diseases, what makes people more vulnerable to outdoor allergens and whether exposure to traffic pollution makes it worse.

We will combine health data from participants of the UK Biobank project with information on daily levels of pollution and allergens in the air that are available across the UK for several years. These environmental data have been developed using statistical models by various research groups in London and abroad. We will also explore new and more informative methods of measuring allergen levels in the air.

The information generated from this project will be useful for government and public health policy makers (support for lowering air pollution, inform how health care utilization should be planned, improve monitoring of allergens in the air), clinicians (improve asthma management and risk factors), as well as patients themselves (understanding risks and how to reduce them). Given that future urbanisation and climate change will lead to important changes in our lifestyles, affect how plants grow and likely lead to higher levels of air pollutants in our cities, it is important that we understand these relationships so that the people in the UK get the right information to protect themselves from asthma attacks.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/determinants-and-health-impact-of-atrial-fibrillation-in-men-and-women

Determinants and health impact of atrial fibrillation in men and women

Last updated:
ID:
9119
Start date:
30 September 2014
Project status:
Current
Principal investigator:
Professor Barbara Casadei
Lead institution:
University of Oxford, Great Britain

To evaluate an algorithm for streamlined detection of rhythm disturbance in resting and exercise one-lead ECG from UKBiobank.

To assess the prevalence and predictors of atrial fibrillation (AF)in the UK middle age population and the impact of this condition on health outcomes in men and women.

To evaluate the determinants of the heart rate response to exercise and post-exercise recovery and the impact of these variables on health outcomes in men and women. The results of this project will generate a gender-specific score for the identification of individuals at a high risk of developing AF and AF-related complications, validate electrocardiographic predictors (such as the abnormal heart rate response to exercise and recovery)of mortality & morbidity and identify the determinants of these parameters in men and women in UKBiobank. We will analyse resting and exercise ECG recordings from the subset of UKBiobank participants who were recruited to this enrichment protocol. ECG parameters such as resting heart rate, exercise heart rate dynamic changes, and rhythm abnormalities will be used. Data on the participants’ medical history, life style, diet, activity data, medications and socioeconomic status as well as the participants’ health outcomes will be analysed both cross-sectionally (based on the initial baseline data) and longitudinally (based on subsequent health outcomes). The study will be conducted in the subset of (ca 120.000) subjects who have one-lead ECG recording during a short bicycle exercise test and recovery and/or at rest at the time of the baseline visit or subsequent repeat-assessment visits.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/determinants-and-predictors-of-individual-differences-in-cognitive-and-brain-aging-from-neurodegenerative-disorders-to-successful-cognitive-aging

Determinants and predictors of individual differences in cognitive and brain aging – from neurodegenerative disorders to successful cognitive aging

Last updated:
ID:
566195
Start date:
26 June 2025
Project status:
Current
Principal investigator:
Dr Sara Pudas
Lead institution:
Umea University, Sweden

Aging-related loss of cognitive and brain function are major contributors to ill health. We will investigate determinants and predictors individual differences in neurocognitive aging, from neurodegenerative disorders to successful neurocognitive aging with minimal loss of function. We will consider blood-based biomarkers such as proteomics, metabolomics, transcriptomics, and telomere length; genetic risk scores; structural and functional neuroimaging measures; health- and lifestyle variables such as hypertension, smoking, and physical activity; as well as psychological traits such as personality. Proxies of biological aging will be computed from omics and clinical markers (e.g., frailty scores). We will account for the heterogenous and multifactorial nature of neurocognitive aging and expect to find subgroups of individuals aging through different mechanisms. Main outcome variables will be neurodegenerative disease status, level and change over time in cognitive function, and MRI-assessed brain characteristics. We will leverage the statistical power of UK Biobank in combination with our local longitudinal datasets for improved characterization of aging trajectories. A large focus will be comparing cross-sectional and longitudinal aging estimates. Another will be to identify early predictors of adverse neurocognitive outcomes. Some analyses will employ the full UK Biobank cohort with available cognitive data to establish normative aging trajectories. Other analyses will be limited to subsets with available data on e.g., neuroimaging and/or omics. The proposed research has the potential to increase knowledge on factors that influence the degree and severity of aging-related cognitive decline in the population, as well as protective factors that can contribute to cognitive resilience. Such factors may also serve as predictive biomarkers that can enable early and individualized interventions. Results will be published in peer-reviewed open-access scientific journals.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/determinants-and-predictors-of-multiple-chronic-diseases-and-traits-identifying-shared-and-unique-molecular-pathways

Determinants and predictors of multiple chronic diseases and traits: identifying shared and unique molecular pathways.

Last updated:
ID:
102297
Start date:
17 May 2023
Project status:
Current
Principal investigator:
Dr Claudia Giambartolomei
Lead institution:
Fondazione Human Technopole, Italy

Complex chronic human disorders such as high blood pressure involve interactions among multiple diseases, risk factors and molecular components that need to be jointly taken into account to (1) gain a better understanding of the biological pathways (2) achieve more accurate predictions of individual risk. Genetic variation within the human genome can be used as an instrument to help decipher shared and unique molecular pathways. For example, genetic variants robustly associated to complex traits and proteomics generate insights on how the genotype could be linked to the phenotype through specific proteins. For the next three years, we aim to dissect differential molecular features across multiple GWAS loci and across multiple diseases. Initially, several separate analyses will independently focus on each disease and condition. We will produce robust predictors of molecular traits across omics studies (e.g. transcriptomic, epigenomic, proteomic and metabolomic markers) and imaging measured together with genetics in the UK biobank population. We will ask, which local and distal genetic effects of molecular intermediates are most predictive of disease? Classical population genetics methods and machine learning (ML) classifiers will be used to provide the best predictors. In a second stage, we will compare findings across multiple diseases and identify pathways affecting the unique or multiple complex diseases. The information from UK biobank allows to identify which components that link to each other at the cellular level show higher comorbidity in the population. We will ask, which genetic effects are more similar/dissimilar across diseases, and determine whether differences in the comorbidity patterns indicate differences in genetic background. We will integrate with drug banks to allow validation with respect to the clinical known drug effects and side-effects. We aim to reproduce the genetic effects on multiple phenotypes, and distinguish the downstream biomarkers known to be affected by the shared pathways. This will allow us to gain a better understanding of treatment of a wide range of illnesses and of multimorbidity. Incorporating information of shared predictors and pathways has the potential to increase risk prediction accuracy; we will explore the benefit to healthcare in treating multiple conditions simultaneously.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/determinants-of-alcohol-consumption-and-its-role-in-health-injury-and-mortality

Determinants of alcohol consumption and its role in health, injury and mortality

Last updated:
ID:
42520
Start date:
10 December 2018
Project status:
Closed
Principal investigator:
Professor Annie Britton
Lead institution:
University College London, Great Britain

At a population level, the burden of harm from drinking alcohol is significant. Using data from UK Biobank, we will investigate the association between alcohol consumption and harm to health. We will consider various aspects of alcohol consumption, including the context of drinking, such as whether it is consumed with meals. Health outcomes will be considered in a broad sense, from injuries to disease and death.

It will also be important to explore possible mechanisms that underlay the relationship between alcohol and health outcomes, so that we can find possible ways to prevent or delay harm. Identifying factors that may lead an individual to drink heavily will inform effective screening and intervention programs to reduce the harm associated with drinking.

To our knowledge this will be the first study to attempt to synthesise these research questions using the unique data in UK Biobank
Given the complexity and the number of variables involved in the analyses we will use, we anticipate that this project will take at least 36 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/determinants-of-cardiorenometabolic-and-vascular-diseases-rare-cancers-and-eye-disease

Determinants of cardiorenometabolic and vascular diseases, rare cancers and eye disease.

Last updated:
ID:
450718
Start date:
3 September 2025
Project status:
Current
Principal investigator:
Dr Daniel Lindholm
Lead institution:
Uppsala University, Sweden

Cardiorenometabolic and vascular diseases remain the most common causes of mortality and are projected to increase globally with an aging population as well as with improved care in developing nations. Eye diseases are common, and visual impairment is an important impediment to self-sufficient and independent living. While several epidemiological studies have established common causes of these diseases, large sample sizes are required to detect uncommon determinants, which, although rare, may improve our knowledge of pathophysiology and lead to more specific treatments.

For more uncommon diseases, including cardiomyopathies, pulmonary hypertension, aortic dissection, but also rare cancers such as small intestinal neuroendocrine tumors, gastric cancer, esophageal cancer, disease determinants are largely unknown. Given their low frequency, large sample sizes and long follow-up time are needed to accurately determine risk factors for disease. This will be a focus of the Swedish Cohorts Consortium, entailing ~1 million participants included from the 1960s onward. Findings from this consortium will need validation in an independent cohort (UK Biobank), and to establish possible causal relationships, Mendelian Randomization will be used.

UK Biobank data will be used both for validation of results from Cohorts.se and other Swedish primary studies, as well as for primary studies in UK Biobank regarding determinants of cardiorenometabolic and vascular diseases, certain cancers and eye disease. In addition to standard methodology, searching for disease determinants in an agnostic way, (e.g. by using AI methods) and finding novel genetic determinants, will provide an opportunity for an unbiased and comprehensive assessment of disease determinants. This has the potential to establish novel and unexpected associations that, if validated, may improve our biological knowledge of these diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/determinants-of-cognitive-and-mental-related-multimorbidity-exploring-joint-associations-of-lifestyle-physical-activity-sedentary-behavior-and-sleep-and-physiological-blood-biomarkers

Determinants of cognitive and mental related multimorbidity: Exploring joint associations of lifestyle (physical activity, sedentary behavior, and sleep) and physiological (blood) biomarkers

Last updated:
ID:
189992
Start date:
17 April 2024
Project status:
Current
Principal investigator:
Professor Tao Huang
Lead institution:
Shanghai Jiao Tong University, China

It’s common of having more than two cognitive and mental related health conditions at the same time, especially among older people, called multimorbidity. This project aims to understand why and how the human lifestyle and blood markers play a joint role in the progression of cognitive and mental related multimorbidity. While there are several previous studies, it remains unclear how physical activity may counteract the adverse effects of sedentary behavior and sleep patterns on cognitive and mental disorders, especially without focusing on multimorbidity. Over the course of three years, the project will analyze data from the UK Biobank, focusing on individuals over 50 with at least two diagnosed cognitive and mental disorders.

The findings could help inform the public about how the daily lifestyle choices may impact their brain and mental health, and hence help them understand what physical activities or habits might help improving cognitive and mental health such as memory loss and depression. Ultimately, the findings are important to help researchers and health professionals design and implement targeted interventions to improve public health and promote healthy ageing.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/determinants-of-disease-progression-and-outcome-in-patients-with-liver-disease

Determinants of Disease Progression and Outcome in Patients With Liver Disease

Last updated:
ID:
105670
Start date:
5 October 2023
Project status:
Current
Principal investigator:
Dr Gautam Mehta
Lead institution:
University College London, Great Britain

Liver disease is a major preventable cause of death in the United Kingdom, and deaths are rising year on year. Since the COVID-19 pandemic began, deaths from liver disease have risen by a further 20% on top of the already high numbers. Most of the deaths, and health care costs, from liver disease occur during the advanced stage called ‘cirrhosis’. Once symptoms occur, this is termed ‘decompensated’ cirrhosis.

The aim of this project is to develop new markers to identify patients who may develop advanced liver disease (cirrhosis), and also those that go on to develop ‘decompensated cirrhosis’. This will allow us to target these patients specifically to prevent the serious complications before they occur. Currently there are no licensed drugs to prevent decompensation, even when liver disease is known about. So, a further important aim of this project is to discover new medicines to prevent liver decompensation occurring.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/determinants-of-eye-diseases

Determinants of eye diseases

Last updated:
ID:
103608
Start date:
9 August 2023
Project status:
Current
Principal investigator:
Dr Alexandre Vallée
Lead institution:
Hôpital Foch, France

Research question: The objective of this project is to investigate social, socio-economic, psychological, and environmental determinants, biological and blood assays, lifestyle behaviors, clinical biomarkers, genetic and radiological features associated with eye physiology, eye impairment, loss of vision and the development of vision diseases in the UK Biobank. We will utilize this database to develop new biomarkers and determinants related to eye diseases.
Scientific rational: As the European population continues to age, the incidence of eye diseases that threaten vision is predicted to steadily rise. These conditions can result in irreversible loss of vision and impaired eyesight, leading to decreased productivity, limitations in performing daily activities, and a significant decrease in quality of life. As a result, healthcare professionals face a significant challenge in addressing this issue both now and in the future. To overcome this challenge, healthcare systems worldwide must move towards a preventive approach, particularly in the field of eye care, where the gap between supply and demand of services is growing globally. To make this shift, it is crucial to gather detailed clinical, biological, social, lifestyle, and imaging data from well-designed population-based cohort studies, such as the UK Biobank. The data provided by the UK Biobank will enable a more comprehensive understanding of the individual and combined effects of various determinants on a range of eye diseases in the adult population. Since these diseases may have multiple causes, each with a modest impact and interacting with others in complex ways, it is crucial to have a large sample size to study the specific effects of each exposure.
However, UK Biobank is not representative of the general population and could not be used to investigate incidence and prevalence of eye diseases but this database could be off interest to better understand the different mechanisms involved and then, the comprehension of eye diseases.
Project duration: 36 months
Public health impact: The results of this project could offer valuable insights into the risk factors for eye diseases that pose a threat to vision. Such insights could enhance our comprehension of the intricate social and environmental mechanisms that damage the development or progression of these diseases and potentially expedite the creation of preventive or early detection programs for significant eye diseases. Additionally, this project can furnish us with useful information on how generalizable the risk factors are and how major eye diseases impact a sizable population.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/determinants-of-functional-decline-multimorbidity-and-neurodegenerative-diseases-a-triangulation-approach

Determinants of functional decline, multimorbidity and neurodegenerative diseases: a triangulation approach

Last updated:
ID:
96856
Start date:
3 March 2023
Project status:
Current
Principal investigator:
Dr Séverine Sabia
Lead institution:
INSERM, France

Longer life expectancy has led to a greater number of people being at risk of functional decline and conditions such as multimorbidity or neurodegenerative disease that increase the risk of disability at older ages. The challenge from a public health perspective is to identify determinants of maintenance of functional capacity as long as possible to reduce the risk and postpone disability onset. In this context, this project aims to identify determinants of functional decline, multimorbidity, and neurodegenerative disease as well as factors involved in progression from these health conditions to death.
Data from the UK Biobank cohort will be used to examine the associations of sociodemographic, behavioural, and health-related factors with change in cognitive, physical, and sensory function over time and risk of onset of multimorbidity, neurodegenerative disease and their progression to death. In order to examine the consistency of findings in different sub-groups, these associations will also be investigated in groups defined by sociodemographic factors (age, sex, ethnicity, education) or health characteristics (obesity, hypertension, diabetes, frailty status). We will also examine the directionality (is it the exposure that influences the health outcome or the reverse?) and the biological plausibility of the associations found. Mendelian randomisation will be used to examine the causal role of exposure in health outcomes. The biological plausibility of the association will be tested by examining whether microvascular function and metabolites play a role in the identified associations.
Taken together, this project will use a comprehensive approach to allow triangulation of findings and provide robust knowledge for future evidence-based personalised prevention interventions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/determinants-of-human-cardiac-structure-and-function-and-relation-with-outcome

Determinants of human cardiac structure and function and relation with outcome

Last updated:
ID:
12010
Start date:
13 April 2015
Project status:
Closed
Principal investigator:
Pim van der Harst
Lead institution:
University Medical Center Groningen, Netherlands

Cardiovascular diseases are a major health burden. Early identification of subjects at risk is important to design strategies to prevent cardiovascular disease or to early intervene. Information of the heart can be derived by functional parameters as well as anatomical (structural) with both provide complementary information and might independently identify subjects at increased risk to experience a cardiovascular event or early death.

Aim:
We aim to study genetic and non-genetic determinants of cardiac function and structure parameters. In addition, we aim to study the independent prognostic value of the cardiac function and structure parameters in predicting clinical outcome. The purpose of UK Biobank is to build a major resource supporting a diverse range of research intended to improve the prevention, diagnosis and treatment of illness and the promotion of health throughout society. This proposal may lead to new understanding of cardiac function, its relation with diseases of the circulatory system and potentially identify new therapeutic targets. In the first phase we will identify and describe the correlates of baseline parameters with parameters of cardiac function as measured by stress testing and of cardiac structure as determined by cardiac MRI. These baseline correlates will be consisting of non-genetic and genetic (GWAS) variables.

In the second phase we will relate the cardiac function and structure parameters with clinical outcomes (cardiovascular events and death) and evaluate the value of these parameters to identify subjects at increased risk. All subjects of whom is available cardiac stress testing and/or cardiac MRI will be included in the analysis.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/determinants-of-human-fat-distribution

Determinants of human fat distribution

Last updated:
ID:
16084
Start date:
1 March 2016
Project status:
Current
Principal investigator:
Professor Fredrik Karpe
Lead institution:
University of Oxford, Great Britain

Obesity is an escalating and significant contributor to mortality and morbidity in the UK and elsewhere. Obesity develops through the imbalance between energy intake and energy expenditure and most research today indicate that central regulation of food intake in a adverse and plentiful environment is the main reason for the imbalance leading to total body fat accumulation. However, the metabolic consequences of regional expansion of body fat depots show a highly interesting and dichotomous pattern: lower body fat accumulation is not associated with increased risk of type 2 diabetes or cardiovascular disease whereas upper body obesity is. There are some indications from the literature that the determinants of regional fat distribution is governed by genes important to tissue development and adipocyte cellularity. Most of this research has been conducted in large datasets but with poor quality assessment of body fat stores, typically waist and hip circumferences measured by tape measure. We intend to use the DXA imaging platforms in the UK Biobank to explore genetic and other determinant of lower vs upper body fat distribution to better understand the mechanisms by which lower body obesity protects against heart disease and type 2 diabetes. This research is in line with the stated aim of the UK Biobank ‘the prevention, diagnosis and treatment of a wide range of life-threatening illnesses’. The first priority is to conduct a non-biased genome-wide search for associations between genotype data and body fat distribution parameters derived from the DXA imaging platform. Other cohorts, such as the Oxford Biobank (n=5,000), have similar measurements and this will support the research.
As a second priority we would want to look for associations between nutritional and social parameters and body fat distribution to lay the foundation for epigenetic mechanisms governing body fat distribution. All participants taking part in the imaging platform. In the first instance we will use data from the 5000 people in the early release and when available analyse the entire data set.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/determinants-of-lung-fibrosis-development-and-progression-in-the-uk-biobank-cohort

Determinants of lung fibrosis development and progression in the UK Biobank cohort

Last updated:
ID:
95988
Start date:
28 November 2023
Project status:
Current
Principal investigator:
Professor Matthew Loxham
Lead institution:
University of Southampton, Great Britain

Idiopathic pulmonary fibrosis (IPF) is a lung disease where the ability of the lungs to allow entry of oxygen into the blood is impaired. No cure is available, and the disease proves fatal, on average 2-4 years after diagnosis. Research has suggested that one trigger for these structural changes may be repeated stress to the lung lining, a source of which could be air pollution. A small number of studies have shown that increased levels of air pollution exposure are associated with a greater risk of developing IPF, and worsening of existing IPF, but findings are inconsistent because of the relatively rare incidence of IPF and difficulties in determining pollution exposure. Based on understanding of how air pollution affects our lungs the cells of our lungs, it is indeed feasible that air pollution exposure may cause IPF, but we do not understand why some individuals may be more at risk than others.

One possible underlying factor may involve telomeres – structures which protect our DNA, but with age become shorter and less effective. Telomeres have been suggested to be shorter in IPF, and may also be shortened by stress to cells. In the whole UK Biobank cohort, there has, perhaps surprisingly, been found to be no association between air pollution exposure and telomere length, but we hypothesise the air pollution exposure may shorten telomeres in a section of the population whose genes render them susceptible, and this may increase the risk of developing IPF.

In this 3-year project, we aim to:
1. Determine whether exposure to air pollution is associated with altered telomere length in a way which is different between individuals with IPF and those who do not have IPF;
2. Determine whether genetic factors associated with maintenance of telomere length or response to air pollution may underlie these differences;
3. Investigate how the observed associations with air pollution might be further influenced by factors relating to lifestyle;
4. Study whether other sources of exposure, such as workplace of domestic, may also be important

The results of this study will help us to understand more about how air pollution exposure might be involved in the development of IPF, as well as improving our knowledge about the processes involved. By studying the role of genetic factors alongside exposures, it may help us identify those most at risk, and thus who would most benefit from interventions to reduce their risk of disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/determinants-of-mitochondrial-dna-mtdna-copy-number

Determinants of mitochondrial DNA (mtDNA) copy number

Last updated:
ID:
24129
Start date:
1 June 2016
Project status:
Current
Principal investigator:
Professor Jonathan Flint
Lead institution:
University of California, Los Angeles, United States of America

We aim to identify what genetic, physiological and metabolic measures mtDNA copy number is associated with, and how this relationship varies with disease. We have previously shown that psychological stress increases the amount of mtDNA. In this project, we will explore how mtDNA variation is determined by nuclear genetic variants and examine its interaction with psychological, physiological and environmental factors. We will focus on medical records and self-reported outcomes of internalising psychiatric disorders and co-morbid health conditions (including autoimmune disorders, like rheumatoid arthritis, and non-immune traits including type2 diabetes, migraine, chronic pain, obesity and body-mass index). This proposed research project investigates the relationship between mtDNA copy number and disease states, and asks how this may be affected by genetic variation. The research will shed light on the link between environmental and physiological stress, cellular metabolism, and diseases (primarily psychiatric illness). It will further the understanding of the mechanisms of these diseases and has the potential to help diagnosis, and potentially guide treatment choice. We will quantify the amount of mtDNA based on the relative genotyping array intensities at mitochondrial probes and nuclear DNA probes. We will then test for associations between the quantified amount of mtDNA and disease status and physiological traits in the dataset to identify relationships between them full cohort


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/determinants-of-sexual-partners-sexual-activity-infectious-diseases-and-diseases-related-to-sexual-behaviors

Determinants of sexual partners, sexual activity, infectious diseases, and diseases related to sexual behaviors.

Last updated:
ID:
101667
Start date:
16 November 2023
Project status:
Current
Principal investigator:
Dr Alexandre Vallée
Lead institution:
Hôpital Foch, France

Stating the aims:
– Determine parameters associated with sexual partners, sexual activity, infectious diseases, and diseases related to sexual behaviors in the UK Biobank middle-aged population.
Scientific rationale:
The lifetime number of different sexual partners is associated with higher risk of sexually transmitted infections. General population is mainly unaware of the risk of sexual transmitted diseases (STIs), as morbidity and mortality risk, except for HIV. Thus, it remains essential to understand the associations between factors (individual, clinical, biological, behavioral) with the increase or decrease in sexual activity and the with STIs and related diseases. The socioeconomic impacts of STIs remained a major public health issue in countries and responsible for around directly attributed 30,000 deaths in United States. Some studies in United States have shown that the number of sexual partners was significantly associated with ethnicity, alcohol, tobacco smoking and dating violence, history of abuse, drugs and violence. Moreover, new mathematical models could explain the relationship between parameters and the number of sexual partners, to better understand the behavior of men and women in order to implement appropriate prevention strategies for safe sex.
Public health impact:
Understanding the determinants associated with sexual partners and sexual activity leading to infectious diseases may be off interest to implement appropriate interventions to educate younger adults about the risk of having a high number of sexual partners with health consequences and thus, the practice of safe sex. Thus, the research projet is to determine determinants associated with sexual partners, sexual activity, infectious diseases, and diseases related to sexual behaviors in the UK Biobank middle-aged population.
Project duration: 36 months


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/determinants-of-somatic-mutation-variability-in-the-uk-biobank-population

Determinants of somatic mutation variability in the UK Biobank population

Last updated:
ID:
50529
Start date:
18 June 2019
Project status:
Closed
Principal investigator:
Dr Gilad Evrony
Lead institution:
NYU Grossman School of Medicine, United States of America

In every population there are families in which many members of the family have cancer. These families usually are found to have mutations in genes that correct or prevent errors in DNA. However, most cancers occur sporadically in individuals in whom their families do not have such a familial predisposition to cancer. Even though these individuals do not come from families with exceptional rates of cancer, they might still have mutations in genes that cause a slightly elevated rather than a highly elevated mutation rate. With the UK Biobank data, we now have an opportunity to detect these slightly elevated levels of DNA mutation in the general population, and what might cause those mutations, to understand why sporadic cancer might occur even when it strikes families without a strong history of cancer. This study might help us eventually be able to predict for each person, many years before cancer might happen, whether they have a slightly higher or lower risk of cancer. Most importantly, this may lead to better approaches for early cancer detection and treatment.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/determinants-of-womens-health-and-cancer-outcomes-a-multi-omics-study-in-the-uk-biobank

Determinants of women’s health and cancer outcomes: a multi-omics study in the UK Biobank

Last updated:
ID:
1079350
Start date:
30 October 2025
Project status:
Current
Principal investigator:
Dr Xin Wu
Lead institution:
Obstetrics and Gynecology Hospital of Fudan University, China

This project aims to investigate determinants of women’s health and cancer outcomes by integrating multi-omics and phenotypic data from the UK Biobank. Women’s health spans endocrine disorders, gynecologic cancers, and psychosocial wellbeing, all of which are critical to quality of life and long-term survival. Yet the biological, lifestyle, and psychosocial factors that shape cancer prognosis and overall health remain incompletely understood.
Our central research questions are: (A)Which multi-omics biomarkers (genomic, transcriptomic, metabolomic) are associated with cancer prognosis and survival in women? (B)How do endocrine, lifestyle, and psychosocial factors interact with molecular features to influence health outcomes? (C)Can integrative multi-omics approaches improve prediction models for cancer outcomes and risk stratification?
The objectives of this study are: (A) To integrate genetic, molecular, lifestyle, and psychosocial data to identify determinants of cancer prognosis and women’s health outcomes. (B) To evaluate prognostic factors and predictive biomarkers that may aid early detection, risk stratification, and personalized management of gynecologic and other cancers. (C) To assess how lifestyle, reproductive history, and psychosocial wellbeing contribute to cancer progression, survivorship, and quality of life.
The rationale lies in the unique opportunity offered by UK Biobank: a large-scale, deeply phenotyped and genotyped cohort with long-term follow-up. By applying multi-omics integration and advanced analytical methods, this research will provide new insights into biological and social determinants of women’s health and cancer outcomes. Ultimately, it will inform public health strategies, support precision medicine for female cancers, and improve wellbeing across the female lifespan.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/determination-of-correlations-between-polygenic-risk-scores-of-40-traits-and-actual-outcomes-using-500-000-individuals

Determination of correlations between polygenic risk scores of 40 traits and actual outcomes using 500.000 individuals

Last updated:
ID:
55495
Start date:
9 December 2019
Project status:
Current
Principal investigator:
Professor Bruce Wolffenbuttel
Lead institution:
University Medical Center Groningen, Netherlands

Scientifically validated methods to calculate the health risks of an individual, such as polygenic risk scores, readily exist, but the public is not benefiting from these as they is not applied beyond research. We will focus on making personalized genomics directly accessible to the public by delivering actionable results, which are aimed at improving their health and lifestyle and ultimately their well-being. We will employ scientific methods to assess the health risks of individuals, based genetic profiling, blood measurements, physical assessment, a questionnaire and activity monitoring using the Fitbit Inspire watch. Based on the individuals health risks, dietary and life style recommendations will be made.

We believe the use of large datasets is the best way to conduct proper research, for which reason the UK Biobank data is uniquely suited for our project. This project specifically entails the risk prediction of 40 traits for 500.000 individuals, such as high cholesterol levels or diabetes, based on genetic profiles and then correlating them to the actual measured values. The strength of that correlation will indicate how well we are able to predict actual outcomes, based on genetic profiles. The outcome will allow us to determine which traits we can and should be using when determining our health recommendations to an individual. Furthermore, we aim to identify lifestyle and dietary changes that could offset the increased disease risk in individuals prone to a disease. These can then be used to advice individuals prone to disease to allow them to live healthier lives.

For example, if turns out that there is a strong correlation between individuals that are diabetic and their genetic risk to become diabetic, we can use this to make health recommendations that reduce the chances of developing diabetes. For example, if an individual is genetically prone to become diabetic and has a high sugar diet, a low sugar diet will be recommended, with the advice to monitor insulin levels on a 3 monthly interval basis. The aim is to prevent disease, rather than curing it, for which purpose the genetic data is uniquely suited.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/determine-genetic-factors-that-modulate-the-alzheimers-disease-risk-effects-of-apoe-4

Determine genetic factors that modulate the Alzheimer’s disease risk effects of APOE-!4

Last updated:
ID:
95743
Start date:
27 September 2023
Project status:
Current
Principal investigator:
Professor Amy Fu
Lead institution:
Hong Kong University of Science and Technology, Hong Kong

Aging-associated diseases have become a leading threat to population health. Of note, Alzheimer’s disease (AD) is one of the most prevalent aging-associated neurodegenerative diseases with no effective intervention strategy.

APOE-!4-a common missense mutation resulting in a mutated protein, is a leading genetic risk factor for many aging-associated diseases including AD. Particularly, individuals harboring APOE-!4 face a 3- to 15-fold higher risk for AD than the general population, and APOE-!4 presents in approximately half of all patients with AD.

However, no appropriate intervention strategies target APOE-!4. Even worse, some intervention strategies exert no effects or even unexpected outcomes in APOE-!4 carriers. Thus, there is an urgent need for developing more accurate genetic test solutions for AD risk forecasting as well as effective AD intervention strategies that specifically target APOE-!4. To accomplish this, a deeper understanding of the effects of APOE-!4 in AD pathogenesis is required.

Identifying factors that modulate APOE-!4 AD risk will shed light on the pathways on lowering APOE-!4 AD risk and boost the development of intervention strategies for APOE-!4 carriers (who are facing a higher AD risk). Recent studies suggested the roles of genetic factors in APOE surrounding regions in modifying APOE-!4 AD risk. While till now, it remained unknown which genetic factors modulate APOE-!4 AD risk and what would be the associated mechanism. Here we initiated a 3-year research project which was recently funded by the General Research Fund at Hong Kong, in which we will try to identify variants that can modify APOE-!4 AD risk, and further investigate their possible effects in modulating the expression of brain APOE/ APOE-!4 transcripts.

This study aimed to: (I) Identify the genomic variants that modify the risk effect of APOE-!4 in Alzheimer’s disease; (II) Examine the effects of candidate variants on modulating APOE expression and Alzheimer’s disease-associated endophenotypes; and (III) Examine the mechanisms of how the candidate variants modify APOE expression.

The successful completion of the captioned project will result in identification of genetic factors that can modify the APOE-!4 AD risk in the general population, with the results will provide a roadmap for an APOE-!4-guided solution for AD diagnosis and therapy, which will enable both a more accurate genetic-based risk assessment for AD, as well as a tailored AD intervention strategy that targets APOE-!4.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/determine-the-association-between-bone-mineral-density-and-the-risk-of-dementia

Determine the association between bone mineral density and the risk of dementia

Last updated:
ID:
199603
Start date:
8 October 2024
Project status:
Current
Principal investigator:
Dr Jun Yuan
Lead institution:
Perron Institute for Neurological and Translational Science, Australia

Aims: Our main goals are to find out if there’s a connection between osteoporosis and the likelihood of experiencing dementia later in life. Additionally, we want to explore whether bone health at different locations in the body (like the spine, hip, and forearm) is linked to the risk of developing late-life dementia. Finally, we’ll check if including bone health data improves our ability to predict dementia accurately.
Scientific rationale: Osteoporosis, a common condition in older individuals, involves lower bone density, raising the risk of fractures in various body parts. Dementia, especially Alzheimer’s disease (AD), affects a significant number of older adults. Both osteoporosis and dementia share increasing age as a major risk factor, and studies suggest a possible connection between bone density and dementia. Some evidence even hints that people with osteoporosis might be more likely to develop AD later in life. However, it’s still unclear if bone density is directly linked to the chances of developing dementia.
Project duration: Our project is expected to take about 3 years from gathering data to obtaining results.
Impact: Dementia is a growing global concern with widespread social and economic effects. Currently, around 50 million people worldwide live with dementia, and this number is predicted to triple by 2050. The challenge is that there can be up to a 20-year period before symptoms appear, making early identification crucial for effective prevention strategies. Our study aims to provide evidence about the relationship between osteoporosis and dementia. Importantly, if our findings show that bone health can predict dementia, it could encourage early lifestyle modifications to support both bone and brain health.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/determine-the-diagnostic-and-prognostic-value-of-circadian-rhythms-and-related-genetic-variations-in-human-diseases

Determine the diagnostic and prognostic value of circadian rhythms and related genetic variations in human diseases

Last updated:
ID:
786804
Start date:
24 June 2025
Project status:
Current
Principal investigator:
Dr Dongyin Guan
Lead institution:
Baylor College of Medicine, United States of America

Patterns of daily human activity are controlled by an intrinsic circadian clock that promotes ~24 hr rhythms in many behavioral and physiological processes. Disruption of the circadian rhythm can cause health conditions in several parts of the body in the long term, such as the cardiovascular and gastrointestinal systems, and is more susceptible to diabetes, obesity, and mental health conditions. Genetic variation is one of the most important factors that influence each person’s circadian rhythm. However, genetic variations associated with circadian rhythm disruption are largely unknown.

Aims In this study, we will determine the role of genetic variations, rhythmic proteins, and metabolites in related diseases.
Research objectives: donors who have genotype, protein, or metabolites abundance data

Study design: We will retrieve the genotype, metabolomic, and proteomic data of thousands of humans and use computing methods in bioinformatics to identify genetic variations that are associated with the destruction of circadian rhythmicity and the role of these variations in related diseases.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/determining-the-carrier-frequency-of-pathogenic-heteroplasmic-mitochondrial-dna-variations-in-the-uk-biobank-cohort-and-their-potential-contribution-to-common-traits

Determining the carrier frequency of pathogenic heteroplasmic mitochondrial DNA variations in the UK Biobank cohort and their potential contribution to common traits.

Last updated:
ID:
40518
Start date:
29 July 2021
Project status:
Current
Principal investigator:
Dr Sarah Jane Pickett
Lead institution:
Newcastle University, Great Britain

Mitochondria are uniquely controlled by both the nuclear genome and their own 16.6kb, circular genome that is present in multiple copies per cell and is maternally-inherited. Pathogenic sequence variation within mtDNA can give rise to mitochondrial diseases, which are a group of disorders that are highly clinically variable and can affect individuals at any age.
It is possible for more than one species of mtDNA molecule to be present within a cell; this is known as heteroplasmy. Many pathogenic mtDNA variants are heteroplasmic meaning that the level of variant present within an individual can range from 0-100%. Gaining an accurate measurement of the level within an individual is important for predicting their likely disease burden. We hypothesise that low levels could be associated with more common clinical phenotypes.
The current computational method for determining genetic variation within UK Biobank samples is tailored towards nuclear DNA variants and not heteroplasmic mtDNA variants. As a result, low levels of pathogenic mtDNA variants may not be detected.
We aim to use the raw fluorescent probe data from the UK Biobank genotyping array and whole exome sequencing data to determine the level of heteroplasmic mtDNA variants within individuals. This will allow us to determine the frequency of pathogenic mtDNA variants in the UK Biobank cohort and to test whether they are associated with phenotypes related to those seen in mitochondrial disease, such as diabetes, deafness, psychiatric disorders, migraine, epilepsy and cardiovascular disease. This will enhance our understanding of the aetiology of associated common phenotypes and allow us to re-evaluate the likely mechanism by which these variants cause mitochondrial disease.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/determining-the-effect-of-immune-mediated-disease-imd-genetic-risk-variants-on-secondary-health-outcomes-and-their-interaction-with-environmental-factors

Determining the effect of immune-mediated disease (IMD) genetic risk variants on secondary health outcomes and their interaction with environmental factors

Last updated:
ID:
10625
Start date:
1 April 2015
Project status:
Closed
Principal investigator:
Professor Lars Fugger
Lead institution:
University of Oxford, Great Britain

Immune-mediated diseases (IMDs), such as multiple sclerosis, comprise a clinically heterogeneous group of disorders affecting ~3-5% of individuals of European ancestry. Our understanding of the exact mechanisms that lead to these conditions remains limited, but scientific progress has shown that both genetic and environmental factors play important roles. We aim to integrate and analyse the extensive genetic, clinical and epidemiological data available through the UK Biobank, in order to identify key shared but also disease-specific mechanisms that may be targeted for therapeutic benefit. One of the aims of the UK Biobank is to improve prevention, diagnosis and treatment of a wide range of illnesses. For many of these illnesses, and in particular for IMDs, genetic and environmental factors play an important role in disease susceptibility. A first step towards meeting this aim for IMDs is to elucidate how these risk factors interact, thereby altering immunological responses and promoting disease development. The proposed research aims to integrate UK Biobank epidemiological, clinical and genetic data to identify parameters for disease risk prediction and diagnosis, and disease mechanisms that may be amenable to drug targeting. Through recent genomic studies an extensive catalogue of genetic variants that influence IMD susceptibility has been constructed. Many of these variants affect risk to multiple conditions, and in some instances have opposing effects on different disorders. For most variants, however, the mechanisms by which they mediate their effects are unknown. The extensive data sets available through the UK Biobank provide a unique opportunity to perform a comprehensive computational assessment of how genetic and environmental factors influence not one but a breadth of different immunological conditions and clinical phenotypes. The results of these analyses will be integrated with laboratory-based studies. In order to perform the required computational analyses and to integrate them with laboratory-based findings, this project requires data regarding lifestyle and epidemiological factors, clinical and para-clinical information, and genetic data. In particular, we are requesting information on all individuals with available genetic data, and with the possibility of updating access to resources, as more individuals are genotyped. For those individuals with available genetic data, we would request access to known diagnoses of IMDs and relevant environmental exposures from the clinical records, such as smocking and known episodes of viral and bacterial infections. No samples are requested.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/determining-the-effects-of-objectively-measured-physical-activity-on-circuit-specific-brain-aging-in-a-massive-multi-modal-neuroimaging-community-sample

Determining the effects of objectively-measured physical activity on circuit-specific brain aging in a massive multi-modal neuroimaging community sample.

Last updated:
ID:
28212
Start date:
4 October 2018
Project status:
Closed
Principal investigator:
Dr Mark Peterson
Lead institution:
University of Michigan, United States of America

Physical activity has beneficial effects on neurocognitive functions and prevention of age-related cognitive declines leading to dementia. Our long-term goal is to better understand the role of exercise on brain structural and functional health through the lifespan. Our objectives are to: (1) determine the effects of objectively-measured physical activity on trajectories of aging of multiple cognition-relevant brain circuits; and (2) parse and quantify mediation pathways between physical activity, circuit-specific brain age, and neurocognitive functioning. The expansion of the aging population, combined with decreasing mortality, has led to a diversification and growth of chronic disease morbidity, including increased prevalence of aging-related mobility and cognitive impairments and a substantial reduction in the number of nondisabled years. Our study is poised to decisively answer 2 critical unanswered questions: (1) What is the effect of physical activity on circuit-specific brain aging? and (2) Does this effect mediate the effect of physical activity on improved cognitive functioning? Our study improves upon existing studies along a number of dimensions. A recent study correlated brain volumes in 331 healthy adults with self-reported measures of physical activity. We will go beyond that study in multiple ways, including: (1) inclusion of over 15x more participants; (2) drawing from a large sample; (3) physical activity will be measured objectively (accelerometry); (4) utilization of multimodal brain imaging; (5) examination of circuit-specific aging trajectories; (6) employment of advanced deep learning methods for constructing brain age trajectories; and finally, (7) we will parse complex relationships between physical activity, brain aging, and cognitive functioning to gain evidence about potential causal pathways. We propose to use imaging from the
subjects as extensively described (Miller KL, et al. Multimodal population brain imaging in the UK Biobank prospective epidemiological study. Nat Neurosci. 2016;19(11):1523-1536), of which over 5,400 have complete neuroimaging, accelerometer data, anthropometric and sociodemographic information, and cognitive functioning data.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/determining-the-genetic-and-environmental-aetiology-of-complex-traits-and-diseases-using-existing-and-novel-statistical-methodologies

Determining the genetic and environmental aetiology of complex traits and diseases using existing and novel statistical methodologies

Last updated:
ID:
53641
Start date:
9 December 2019
Project status:
Current
Principal investigator:
Professor David Evans
Lead institution:
University of Queensland, Australia

Complex traits are a result of many different genetic and environmental factors. However, for most traits, the identity of these factors are unknown. Understanding the genes and environmental factors that underlie complex traits is useful in terms of elucidating the biological processes underlying complex phenotypes, and in the case of common diseases and medically relevant traits, can lead to the identification of novel drug targets and the development of public health strategies to decrease future risk of pathology. Our overall aim is to understand the genetic and environmental aetiology of complex traits and diseases. Specifically we will develop and apply novel and existing statistical genetics methodologies to (a) understand the genetic and environmental aetiology of complex traits and diseases, (b) understand the source and structure of the correlation between different complex traits and diseases, and (c) to estimate the causal relationship between different complex traits and diseases. In order to achieve our aim we will (a) analyse data from the UK biobank and combine the results with other genetics and -omics data sets that we have in house, and (b) develop and apply novel and existing statistical genetics methods to these data.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/determining-the-genetic-basis-of-rotator-cuff-tearing-and-arthritis

Determining the Genetic Basis of Rotator Cuff Tearing and Arthritis

Last updated:
ID:
717667
Start date:
23 July 2025
Project status:
Current
Principal investigator:
Dr Robert Tashjian
Lead institution:
University of Utah, United States of America

Our purpose is to identify genetic variants associated with rotator cuff tearing and glenohumeral arthritis. Aim1 – Identify patients from the UK Biobank with rotator cuff tears based upon ICD9 and ICD 10 codes. Aim 2- Identify patient from the UK Biobank with primary glenohumeral osteoarthritis based upon ICD9 (715.11) and ICD 10 codes (M19.011- left shoulder, m19.012- right shoulder, and M19.019- unspecified shoulder) Aim 3- Query UK Biobank for xrays minimum 2 views of shoulder (73030) Aim 4- Perform genetic analysis estimating the risk of disease for rare shared haplotypes by comparing all patients with rotator cuff tears and primary glenohumeral osteoarthritis compared to all other patients in the UK Biobank. This method will allow identification of variants associated with rotator cuff tearing and osteoarthritis.

Rotator cuff disease affects over 17 million U.S. people accounting for over 3.8 million physician visits per year. Despite its widespread prevalence, very little is known regarding its genetic basis or heritability. We have also identified various genetic variants associated with the predisposition to rotator cuff tearing including SNPs in ESRRB, SASH1 and SAP30BP genes. (Tashjian JSES 2015, Tashjian JSES 2016) Identification of further variants is limited due to small sample sizes. We have developed a methodology to identify further variants in small sample sets using share genetic segments between individuals to allow identification of familial relationships. There is currently no data on the genetic etiology of glenohumeral osteoarthritis


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/determining-the-genetic-basis-of-rotator-cuff-tearing

Determining the Genetic Basis of Rotator Cuff Tearing

Last updated:
ID:
47706
Start date:
4 June 2019
Project status:
Closed
Principal investigator:
Dr Robert Tashjian
Lead institution:
University of Utah, United States of America

Rotator cuff tearing is a common shoulder problem. The development of tearing is likely due to environmental and genetic factors. Identifying genetic variants associated with rotator cuff tearing will allow physicians to identify patients at risk for genetic component to tearing. These patients are likely to have worse healing as well after repair and therefore may help indications for treatment. Our aims include identifying patients from the UK biobank with rotator cuff tears and then do genetic analyses to compare these patients to a control cohort from the population. Identification of variants will improve our ability to indicate patients for treatment. The duration is expected to be 6 months. The health impact is large as millions of people are effected by rotator cuff tearing and improving the ability to properly treat them is critical. Genetic factors leading to tearing will be a component of indicating these patients.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/determining-the-mechanisms-by-which-exercise-affects-cancer

Determining the mechanisms by which exercise affects cancer

Last updated:
ID:
91866
Start date:
4 December 2024
Project status:
Current
Principal investigator:
Dr Paul Christopher Boutros
Lead institution:
University of California, Los Angeles, United States of America

An estimated two-thirds of cancer deaths in the U.S. can be attributed to modifiable risk factors such as smoking, diet and exercise. Exercise has been shown to exert anti-tumor effects, including the reduction of incidence of cancer, to provide benefit as adjunct therapy during cancer treatment and to improve clinical outcomes by decreasing recurrence and mortality when occurring after diagnosis. While exercise is the strongest positive modifiable risk factor, surprisingly, the molecular mechanisms by which it influences cancer evolution are still almost entirely unknown. To address this gap in knowledge, we aim to enhance the molecular understanding of exercise oncology by precisely identifying clinical and molecular features associated with exercise. We will characterize the effects of exercise on cancer mutational and evolutionary processes (Aim 1), assess host biomarker responses to exercise (Aim 2), and evaluate the role of genetics and dose-dependent exercise interactions in cancer-related outcomes (Aim 3). The UKBB is a unique resource that provides physical activity measurements, genomic profiling, and clinical outcomes for a large cohort of human individuals. Exercise therapy has significant public health potential due to its accessibility, safety, cost effectiveness and ability to give individuals agency to positively influence their own risk of cancer or clinical outcomes. We hope our work will reveal important insights into the mechanisms by why exercise affects cancer and viability of prescribing exercise therapy as a preventative, adjunct and treatment strategy.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/determining-the-outcomes-of-people-with-liver-disease

Determining the Outcomes of People with Liver Disease

Last updated:
ID:
9914
Start date:
1 November 2015
Project status:
Current
Principal investigator:
Dr Rajarshi Banerjee
Lead institution:
Perspectum Diagnostics Ltd, Great Britain

Perspectum Diagnostics has developed a method of analysing magnetic resonance imaging (MRI) data that gives an accurate estimate of the amount of liver fat, the amount of liver iron, and the extent of inflammation and scarring in the liver. These three characteristics of the liver are also the most important in the diagnosis of liver disease. By analysing the abdominal MR images from all UK Biobank participants, we can determine approximately how many have abnormal liver composition, and the distribution of each of these measures in the population. Finally, and most importantly, we can examine the outcomes of the participants with liver disease, and determine which biomarkers are predictive of these outcomes. New, clinically meaningful data will be generated from the existing DICOM images, and fed back in to the UK Biobank data repository. These data will be directly relevant to future health outcomes and of use to other researchers. Excess liver fat is associated with coronary artery atheroma and metabolic syndrome, and is strongly associated with obesity-related disease. Liver fibrosis and inflammation are both associated with adverse outcomes, which is especially relevant in those with fatty liver disease. We will be able to show which patients have liver disease, and future researchers can link these findings to specific outcomes. The MRI scans from the imaging enhancement study will be analysed by LiverMultiScan to determine liver fat, iron, inflammation and fibrosis (LIF score). These measures have separately been validated against liver biopsies from patients.

These data will then be compared to measures of body composition, serum markers (lipid profile, iron stores, CRP and others) and habits associated with liver disease (eg alcohol intake, exercise and diet).

We will follow up all patients and identify those with a liver-related clinical outcome (eg liver failure, hepatic encephalopathy), and determine which prognostic factors best predict these outcomes in this population.
All 100,000 participants from the UK Biobank imaging enhancement study (ie the full cohort from the imaging enhancement study) will be analysed to determine the baseline liver health profiles of the population.

Clinical outcomes data will be collected, with the aim of capturing
– every liver-related death
– every episode of oesophageal variceal bleeding
– every new diagnosis of cirrhosis
– every new diagnosis of liver failure or gross ascites due to liver disease (excluding malignant ascites)
– every new primary hepatocellular carcinoma and cholangiocarcinoma
– every new pancreatic carcinoma


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/determining-trajectory-of-menstrual-cycle-by-developing-verifying-and-validating-an-artificial-intelligence-based-digital-twin-for-heavy-menstrual-bleeding-hmb

Determining Trajectory of Menstrual Cycle by Developing, Verifying, and Validating an Artificial Intelligence-based Digital Twin for Heavy Menstrual Bleeding (HMB)

Last updated:
ID:
886309
Start date:
23 July 2025
Project status:
Current
Principal investigator:
Dr Vinay Pai
Lead institution:
Health Tequity, LLC., United States of America

In this project, we aim to develop trajectories of the menstrual cycle by constructing artificial intelligence based digital twins of heavy menstrual bleeding.
Digital twins (DT) potentially offer a model that leverages population level data and combines it with the woman’s unique data, to deliver personalized recommendations for self-management and treatment/care plans to ensure early identification of HMB onset and can be scalable across the global space. The two-way dynamic flow of information between the physical and virtual world (the twinning) can yield more accurate digital representations of the health conditions and enable enhanced in-depth virtual testing of potential behavioral, social, and/or medical interventions.
We will generate a menstrual cycle DT via deep phenotyping, or the integration and automated processing of data from a wide range of factors related to the menstrual cycle. Some of the menstrual factors included (as available) pregnancy test, complete blood count including hemoglobin, serum iron, ferritin or total iron-binding capacity (TIBC), hormonal profile to include thyroid function tests, follicle-stimulating hormone (FSH), luteinizing hormone, estrogen, progesterone, testosterone. Additionally, we would also need information (as available) such as age at menarche, age at menopause, length of menstrual cycle, use of hormone-replacement therapy, use of oral contraceptive pills, etc.
We will approach DT generation using two different AI methods: first, a generative model using variational autoencoder (VAE) and second, using an agentic-AI approach coupling a large language model (LLM) with retrieval-augmented generation, and incorporate causal analysis and uncertainty estimation to determine statistical power. For verification and validation of the digital twins, we will combine the UK Biobank data with data from additional sources such as from the Institute for Health Metrics (IHM, USA), and the AllofUs project (NIH, USA).


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/develop-an-in-house-wgs-solution-for-exploring-rare-disease-insights

Develop an in-house WGS solution for exploring rare disease insights

Last updated:
ID:
91144
Start date:
5 April 2023
Project status:
Current
Principal investigator:
Dr Pascal-Antoine Christin
Lead institution:
SOPHiA GENETICS, Switzerland

To develop a solution for the exploration of rare diseases on the whole genome of a patient, we need to have benchmark data for testing the efficacy of our solution. As rare diseases are complex – finding the genetic needle in the haystack – having a solution that will explore the entire human genome, both the coding and the non-coding component will bring us closer to solving this complex puzzle.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/develop-biomarker-of-chronic-fatigue-syndrome-cfs-from-accelerometer-data

Develop biomarker of chronic fatigue syndrome (CFS) from accelerometer data

Last updated:
ID:
41746
Start date:
19 December 2018
Project status:
Closed
Principal investigator:
Dr Vladimir Morozov
Lead institution:
Shire Pharmaceuticals LLC, United States of America

Chronic fatigue syndrome/myalgic encephalomyelitis (CFS/ME) is a complex disease that can be debilitating. The exact cause or causes of CFS/ME are unknown. Symptoms affect several body systems and may include severe fatigue or exhaustion, unrefreshing sleep, weakness, muscle and joint pain, impaired memory or concentration. ME/CFS symptoms may get worse after any activity, whether it’s physical or cognitive. There are no approved therapies indicated to treat CFS/ME. Development of new CFS/ME treatments is complicated by a lack of understanding of how the disease develops and its lengthy diagnostic path. Indeed, because of multiple symptoms, ME/CFS diagnosis is challenging. In order to better understand the impact of the disease on activity and in particular the post -exertional fatigue that is related to activity, we would like to develop a computer algorithm that translates accelerometer data into measure of CFS/ME severity. We call it digital biomarker. Such a digital biomarker would allow diagnosis and monitoring of CFS/ME patients remotely, cheaply and non-invasively. We are going to test the accelerometer biomarker along with patient-reported outcomes in a small study this year. If it is proven to be accurate, we will use this biomarker in bigger clinical trials testing new treatments for CFS/ME.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/develop-multi-modality-medical-qa-question-answering-systems

Develop multi-modality medical QA (Question Answering) systems

Last updated:
ID:
106456
Start date:
20 March 2024
Project status:
Current
Principal investigator:
Mr Chaoyi Wu
Lead institution:
Shanghai Jiao Tong University, China

Our study aims to enhance the precision, adaptability, and usefulness of artificial intelligence models in the medical field. We plan to develop a comprehensive multimodal foundation model infused with medical-specific knowledge to create a general Medical Question Answering system. This system will be capable of quickly answering medical questions based on user’s input in any format, such as text or images, facilitating convenient access to top-notch medical resources for healthcare centers and online services. By utilizing advanced deep learning techniques and leveraging data from the biobank, we aim to improve public health by providing faster and more accessible medical services, including online diagnosis, treatment advice, and medication recommendations. This ambitious project is expected to span 36 months.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/develop-statistical-genetic-methods-with-hidden-subgroups

develop statistical genetic methods with hidden subgroups

Last updated:
ID:
78898
Start date:
26 January 2023
Project status:
Current
Principal investigator:
Professor Wei Li
Lead institution:
University of California, Irvine, United States of America

In this study, we will develop novel methods to identify who (which subgroup of people) have a higher risk of diseases (e.g., breast cancer) than others.

Previously, scientists have shown that some humans are born with variations in their DNA (known as “risk variants”) that increase their lifetime risk of some diseases (e.g., breast cancer). Using these risk variants, scientists defined the polygenic risk score (PRS) to predict if a person will have a high risk to have a certain disease (e.g., breast cancer). However, previous studies do not consider the difference between people. In this study, we will develop novel methods, which will take into account population differences, to obtain more accurate disease risk predictions.

We will mainly focus on brain diseases and breast cancer first. And if promising, our methods will be extended to other human diseases. The whole project is expected to last for about three years.

We believe that our study will make significant contributions to disease (e.g., breast cancer and brain diseases) risk prediction. From a public health perspective, if we can identify the subgroup of people who have high disease risk, then we can provide appropriate care for them.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/developing-a-bayesian-methodology-for-identifying-disease-associated-dysregulation-of-genes-and-gene-subnetworks-by-integration-of-gwas-and-eqtl-data-with-application-to-cardiovascular-diseases

Developing a Bayesian methodology for identifying disease-associated dysregulation of genes and gene subnetworks by integration of GWAS and eQTL data, with application to cardiovascular diseases.

Last updated:
ID:
31314
Start date:
27 March 2018
Project status:
Closed
Principal investigator:
Dr Johannes Soeding
Lead institution:
Max Planck Institute for Biophysical Chemistry, Germany

We aim to develop a Bayesian methodology for identifying disease-associated dysregulation of genes and gene subnetworks by integrating GWAS and eQTL data. This will improve our understanding of the underlying mechanism of the common, non-infectious diseases. The genes and gene modules will represent prime targets for pharmacological intervention. We plan to use phenotypes related to cardiovascular diseases (CVD) to validate our method. Our software will produce ranked list of genes and gene modules which are statistically associated to diseases. These will have clinical utility for prognosis or treatment. The proposed method will also improve the prediction of disease propensity of any individual given his genotype. Hence it can be used for improved diagnosis and prevention of common, non-infectious diseases. We have formulated the theory for the proposed method, and we are in the process of developing a software tool. We will find CVD cases and matched controls from the UK Biobank data, and apply our method to discover genes and gene subnetworks whose dysregulation is statistically coupled to increased risk for CVD. We would like to use the largest sample size available to maximize the power of detecting statistical associations.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/developing-a-diagnostic-tool-for-non-alcoholic-fatty-liver-disease-and-inferring-causal-relationships-with-type-2-diabetes

Developing a diagnostic tool for non-alcoholic fatty liver disease and inferring causal relationships with type 2 diabetes

Last updated:
ID:
18274
Start date:
4 February 2019
Project status:
Closed
Principal investigator:
Professor Paul Franks
Lead institution:
Lund University, Sweden

Non-alcoholic fatty liver disease (NAFLD) and Type 2 diabetes (T2D) are two pervasive and globally prevalent diseases that cause serious health complications. Utilizing UKBB rich dataset we aim to investigate two key questions regarding these diseases. In “Question 1”, we aim to derive a prediction tool for the diagnosis of NAFLD and in “Question 2” our aim is to assess the causal pathways between NAFLD and T2D.

With respect to “Question 1”, the diagnosis of NAFLD at an early stage is of great importance as this can help us to go with interventions and prevent further damages to the liver. Liver biopsy, MRI scans, ultrasounds and liver enzyme tests are often used for the diagnosis of the disease. However, the invasive nature of the liver biopsy, the high costs of the MRI scans and lack of accuracy with only relying on liver enzyme tests leave many patients with NAFLD undiagnosed. With developing a prediction tool for NAFLD, we aim to introduce a strong proxy as a safe and cheaper alternative to MRI scans and limit the number of MRI scans performed only to those most likely to have fatty liver.

In “question 2”, our aim is investigating the causal relationship between NAFLD and T2D. Many studies show the association between these diseases, but whether they are causally related and if so what the direction and magnitude of this effect are is not clear. Determining the cause-effect relationships between T2D and NAFLD may help us with the design of interventions for their prevention and treatment. Within the project we will build a structural graph including these disease and several other clinical factors to further explore the causal pathways that connect them together.

These two questions are implicitly related to each other. Using the prediction model from “question 1” people at risk of NAFLD can be identified, who might subsequently be intervened upon. However, one would not want to intervene unless the relationship between liver fat and disease (diabetes in this case) is causal, which we will try to address in “question 2”.

Our estimated timeline for the proposed research project in total is 24 months. Which comprises of exploring data and finalizing aforementioned methods in our analysis (6months), applying methods on the data (12months) and drafting/submitting two papers from our analyses to a relevant scientific journals (6months).


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/developing-a-look-a-like-model-for-the-prediction-of-premature-mortality-and-chronic-disease

Developing a look a like model for the prediction of premature mortality and chronic disease

Last updated:
ID:
54045
Start date:
30 October 2019
Project status:
Closed
Principal investigator:
Dr Filippo Menolascina
Lead institution:
University of Edinburgh, Great Britain

Identifying patients mortality risks and in particular the impact of chronic disease is key to preventative medicine. In recent years a number of machine learning algorithms have been applied to this problem. In this study, we will attempt to validate look-a-like modelling as an alternative approach. Look-a-like modelling has been hugely successful in other industries. It enables new inputs to be categorised based on their similarities to known positive examples. Unlike other machine learning models, look-a-like models are not considered black boxes (that is to say they can be easily interpreted). There is every reason to believe that they should be able to identify patients at risk of premature death or chronic disease. We propose a systematic comparison of look-a-like modelling and machine learning approaches to risk prediction (Deep learning, Random Forest and Cox regression).

Machine learning techniques have already achieved superhuman performance in many fields of diagnostic medicine, De Fauw et al (2018). The continual evaluation of novel approaches is essential to this process. Accurate risk prediction algorithms should facilitate early and effective interventions, when treating disease.

We require access to clinical, medical history and sociodemographic data from the UK Biobank. We will then perform feature selection using the predictive potential of these data, in particular with regard to specific disease onset. To evaluate these models predictive capabilities we would require access to the full UK Biobank cohort (approx. n=500,000). This would enable us to develop robust models which could be validated on previously unseen data (essential in machine learning).


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/developing-a-multi-ethnic-disease-risk-prediction-model-enhancing-generalizability-across-diverse-populations-through-genomic-and-machine-learning-approaches

Developing a Multi-Ethnic Disease Risk Prediction Model: Enhancing Generalizability Across Diverse Populations Through Genomic and Machine Learning Approaches

Last updated:
ID:
733838
Start date:
25 July 2025
Project status:
Current
Principal investigator:
Dr Yunsung Cho
Lead institution:
CG Invites Co., Ltd., Korea (South)

Questions: How can we develop a multi-ethnic disease risk prediction model that enhances generalizability across diverse populations?

Objective: We aim to develop and validate a disease risk score model with improved generalizability across diverse populations. Specifically, this project seeks to develop a generalized multi-ethnic disease risk prediction model that can accurately assess disease risk across different ethnic groups.

Scientific rationale: The development of generalizable disease risk prediction models has become increasingly important due to rising global migration, which has led to a decline in single-ethnicity populations and the emergence of genetically diverse communities. While existing ethnicity-specific models may perform well within their respective populations, their accuracy in admixed or multi-ethnic groups remains uncertain, limiting broader applicability.
To address this, we aim to develop a multi-ethnic disease risk prediction model using UK Biobank data alongside our machine learning models trained on 80,000 Korean individuals, covering 39 diseases in males and 41 in females, with an average of 50 disease-specific markers per disease.
As part of risk model construction, we will integrate comparative genomics to characterize subpopulation genetic structures and assess their disease susceptibilities. Given that most of our existing data originates from Jeju Island, we aim to characterize its genetic distinctiveness compared to the Korean mainland. Through population genetics analyses, we will extract genetic parameters to refine the model, ensuring population structure and genetic diversity are adequately represented in disease risk predictions.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/developing-a-reference-normative-sample-of-the-volumetric-and-myelin-density-of-the-spinal-cord-and-brain-imaging-data-in-the-uk-biobank

Developing a reference normative sample of the volumetric and myelin density of the spinal cord and brain imaging data in the UK Biobank

Last updated:
ID:
47233
Start date:
26 March 2019
Project status:
Closed
Principal investigator:
Dr Arman Eshaghi
Lead institution:
University College London, Great Britain

Patients with multiple sclerosis have a relentless shrinkage of the brain and spinal cord. Researchers rely on measurements from healthy volunteers to precisely detect when patients show abnormality. However, even after precisely measuring the change in brain and spinal cord scans of patients with multiple sclerosis, it is still a challenge to provide personalised prediction of the current and future progression of a patient. This is in part due to the lack of reference measures that can provide a normative sample of the change in the brain and spinal cord. We are in the process of processing more than 25,000 MRI scans from patients with MS. Here, I aim to leverage the large dataset available as part of the UK Biobank to construct normative samples which will later be used to stage and predict future progression of MS in patients.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/developing-a-risk-prediction-model-for-chronic-disease-mortality-using-multi-level-survival-modelling

Developing a risk prediction model for chronic disease mortality using multi-level survival modelling

Last updated:
ID:
24321
Start date:
15 December 2016
Project status:
Closed
Principal investigator:
Mr Paul Nash
Lead institution:
Road to Health Group Limited, Great Britain

Health risk assessment of future disease has a number of applications and is in widespread use among clinicians and the general public. However, most applications have been focussed on single disease areas. Predicting death from diverse disease outcomes is complex and influenced by environmental and individual characteristics. In this study, we will explore the relationship between both environmental and individual characteristics and death due to major chronic diseases (cardiovascular, diabetes, pulmonary, dementia, cancers). This will lead to developing a novel and holistic application which can predict risk of death from chronic disease, informing individuals to take preventive measures. The study proposes to use novel methodology to: (i) identify the important social and clinical determinants of chronic disease mortality, and (ii) develop and validate a new prediction model for future mortality risk. By using a current and large population based cohort, we will be able to generate findings which will inform clinicians, policy-makers, and the general public of the most pertinent determinants of health. In developing and validating a risk prediction model, we can also generate useful applications for the prediction of an individual’s future risk of death. The UK Biobank study cohort will be interrogated for demographic, clinical, medical history, family history, prescribing and behavioural variables, to develop an individual patient level predictive model in the first phase. These variables will then be assessed for their relationship with chronic disease mortality. In the second phase, post-code and regional data will be added into the model to create a `multi-level model` which accounts for the clustering of patients with similar individual characteristics in relation to the area of residence. This model will then be tested on whether it predicts mortality accurately or not.
Due to developing a holistic model of mortality which will require investigation and assessment of multiple determinants of health, we will require the full cohort (approx. n = 500,000). To develop and validate the novel risk model will require splitting the dataset into a 75% random sample to develop the prediction model and the remaining 25% sample to validate the model. The use of the full cohort provides adequate sample size for this procedure, and will result in the development of a robust model.


(Internet Archive copy)


https://www.ukbiobank.ac.uk/projects/developing-a-risk-score-for-prediction-of-cardiovascular-disease-in-patients-with-chronic-neurological-diseases-based-on-traditional-and-novel-risk-factors

Developing a risk score for prediction of cardiovascular disease in patients with chronic neurological diseases based on traditional and novel risk factors

Last updated:
ID:
45291
Start date:
25 January 2019
Project status:
Closed
Principal investigator:
Dr Mahima Kapoor
Lead institution:
University College London, Great Britain

Cardiovascular disease, such as heart attacks and strokes are the most common cause of early death in the UK, and are caused by a complex interaction between different risk factors such as age, sex, cholesterol, diabetes and smoking. There are excellent scoring systems available to general practitioners to measure an individual’s risk and then treat identified risk factors to prolong patients’ cardiovascular-disease free years. These scores cannot be directly applied to patients with neurological conditions because other factors such as difficulty walking and the treatment they are on might also influence their individual risk of having a heart attack or stroke. One of the treatments we are specifically interested in investigating is intravenous immunoglobulin (IVIg), a blood product given regularly to patients with inflammatory, neurological conditions to dampen down inflammation attacking nerves and muscles. This treatment is given regularly over many years and it protects the nerves and muscles from damage, prevents disability and maintains independ