1
|
Hartwell EE, Jinwala Z, Milone J, Ramirez S, Gelernter J, Kranzler HR, Kember RL. Application of polygenic scores to a deeply phenotyped sample enriched for substance use disorders reveals extensive pleiotropy with psychiatric and somatic traits. Neuropsychopharmacology 2024; 49:1958-1967. [PMID: 39043921 PMCID: PMC11480112 DOI: 10.1038/s41386-024-01922-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 06/07/2024] [Accepted: 06/28/2024] [Indexed: 07/25/2024]
Abstract
Co-occurring psychiatric, medical, and substance use disorders (SUDs) are common, but the complex pathways leading to such comorbidities are poorly understood. A greater understanding of genetic influences on this phenomenon could inform precision medicine efforts. We used the Yale-Penn dataset, a cross-sectional sample enriched for individuals with SUDs, to examine pleiotropic effects of genetic liability for psychiatric and somatic traits. Participants completed an in-depth interview that provides information on demographics, environment, medical illnesses, and psychiatric and SUDs. Polygenic scores (PGS) for psychiatric disorders and somatic traits were calculated in European-ancestry (EUR; n = 5691) participants and, when discovery datasets were available, for African-ancestry (AFR; n = 4918) participants. Phenome-wide association studies (PheWAS) were then conducted. In AFR participants, the only PGS with significant associations was bipolar disorder (BD), all of which were with substance use phenotypes. In EUR participants, PGS for major depressive disorder (MDD), generalized anxiety disorder (GAD), post-traumatic stress disorder (PTSD), schizophrenia (SCZ), body mass index (BMI), coronary artery disease (CAD), and type 2 diabetes (T2D) all showed significant associations, the majority of which were with phenotypes in the substance use categories. For instance, PGSMDD was associated with over 200 phenotypes, 15 of which were depression-related (e.g., depression criterion count), 55 of which were other psychiatric phenotypes, and 126 of which were substance use phenotypes; and PGSBMI was associated with 138 phenotypes, 105 of which were substance related. Genetic liability for psychiatric and somatic traits is associated with numerous phenotypes across multiple categories, indicative of the broad genetic liability of these traits.
Collapse
Affiliation(s)
- Emily E Hartwell
- Crescenz VA Medical Center, Philadelphia, PA, USA
- University of Pennsylvania, Philadelphia, PA, USA
| | - Zeal Jinwala
- Crescenz VA Medical Center, Philadelphia, PA, USA
- University of Pennsylvania, Philadelphia, PA, USA
| | | | | | - Joel Gelernter
- West Haven VA Medical Center, West Haven, CT, USA
- Yale University, New Haven, CT, USA
| | - Henry R Kranzler
- Crescenz VA Medical Center, Philadelphia, PA, USA
- University of Pennsylvania, Philadelphia, PA, USA
| | - Rachel L Kember
- Crescenz VA Medical Center, Philadelphia, PA, USA.
- University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
2
|
Misra A, Truong B, Urbut SM, Sui Y, Fahed AC, Smoller JW, Patel AP, Natarajan P. Instability of high polygenic risk classification and mitigation by integrative scoring. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.24.24310897. [PMID: 39211853 PMCID: PMC11361234 DOI: 10.1101/2024.07.24.24310897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Polygenic risk scores (PRS) continue to improve with novel methods and expanding genome-wide association studies. Healthcare and commercial laboratories are increasingly deploying PRS reports to patients, but it is unknown how the classification of high polygenic risk changes across individual PRS. Here, we assessed association and classification performance of cataloged PRS for three complex traits. We chronologically ordered all trait-related publications (Pub n ) and identified the single PRS Best(Pub n ) for each Pub n that had the strongest association with the target outcome. While each Best(Pub n ) demonstrated generally consistent population-level strengths of associations, classification of individuals in the top 10% of each Best(Pub n ) distribution varied widely. Using the PRSmix framework, which integrates information across several PRS to improve prediction, we generate corresponding ChronoAdd(Pub n ) scores for each Pub n that combine all polygenic scores from all publications up to and including Pub n . When compared with Best(Pub n ), ChronoAdd(Pub n ) scores demonstrated more consistent high-risk classification amongst themselves. This integrative scoring approach provides stable and reliable classification of high-risk individuals, and is an adaptable framework into which new scores can be incorporated as they are introduced, integrating easily with current PRS implementation strategies.
Collapse
|
3
|
Mekhael M, Bidaoui G, Falloon A, Pandey AC. Personalization of primary prevention: Exploring the role of coronary artery calcium and polygenic risk score in cardiovascular diseases. Trends Cardiovasc Med 2024:S1050-1738(24)00094-X. [PMID: 39442739 DOI: 10.1016/j.tcm.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/14/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
Personalized healthcare is becoming increasingly popular given the vast heterogeneity in disease manifestation between individuals. Many commonly encountered diseases within cardiology are multifactorial in nature and disease progression and response is often variable due to environmental and genetic factors influencing disease states. This makes accurate early identification and primary prevention difficult in certain populations, especially young patients with limited Atherosclerotic Cardiovascular Disease (ASCVD) risk factors. Newer strategies, such as coronary artery calcium (CAC) scans and polygenic risk scores (PRS), are being implemented to aid in the detection of subclinical disease and heritable risk, respectively. Data surrounding CAC scans have shown promising results in their ability to detect subclinical atherosclerosis and predict the risk of future coronary events, especially at the extremes; however, predictive variability exists among different patient populations, limiting the test's specificity. Furthermore, relying only on CAC scores and ASCVD risk scores may fail to identify a large group of patients needing primary prevention who lack subclinical disease and traditional risk factors, but harbor genetic variabilities strongly associated with certain cardiovascular diseases. PRS can overcome these limitations. These scores can be measured in individuals as early as birth to identify genetic variants placing them at elevated risk for developing cardiovascular disease, irrespective of their current cardiovascular health status. By applying PRS alongside CAC scores, previously overlooked patient populations can be identified and begin primary prevention strategies early to achieve optimal outcomes. In this review, we expand on the current knowledge surrounding CAC scores and PRS and highlight the future possibilities of these technologies for preventive cardiology.
Collapse
Affiliation(s)
- Mario Mekhael
- Section of Cardiology, Deming Dept of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Ghassan Bidaoui
- Section of Cardiology, Deming Dept of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Austin Falloon
- Section of Cardiology, Deming Dept of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Amitabh C Pandey
- Section of Cardiology, Deming Dept of Medicine, Tulane University School of Medicine, New Orleans, LA, United States; Southeast Louisiana Veterans Health Care System, New Orleans, LA, United States.
| |
Collapse
|
4
|
Abramowitz SA, Boulier K, Keat K, Cardone KM, Shivakumar M, DePaolo J, Judy R, Kim D, Rader DJ, Ritchie MD, Voight BF, Pasaniuc B, Levin MG, Damrauer SM. Population Performance and Individual Agreement of Coronary Artery Disease Polygenic Risk Scores. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.25.24310931. [PMID: 39108513 PMCID: PMC11302700 DOI: 10.1101/2024.07.25.24310931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/12/2024]
Abstract
Importance Polygenic risk scores (PRSs) for coronary artery disease (CAD) are a growing clinical and commercial reality. Whether existing scores provide similar individual-level assessments of disease liability is a critical consideration for clinical implementation that remains uncharacterized. Objective Characterize the reliability of CAD PRSs that perform equivalently at the population level at predicting individual-level risk. Design Cross-sectional Study. Setting All of Us Research Program (AOU), Penn Medicine Biobank (PMBB), and UCLA ATLAS Precision Health Biobank. Participants Volunteers of diverse genetic backgrounds enrolled in AOU, PMBB, and UCLA with available electronic health record and genotyping data. Exposures Polygenic risk for CAD from previously published PRSs and new PRSs developed separately from the testing cohorts. Main Outcomes and Measures Sets of CAD PRSs that perform population prediction equivalently were identified by comparing calibration and discrimination (Brier score and AUROC) of generalized linear models of prevalent CAD using Bayesian analysis of variance. Among equivalently performing scores, individual-level agreement between risk estimates was tested with intraclass correlation (ICC) and Light's Kappa, measures of inter-rater reliability. Results 50 PRSs were calculated for 171,095 AOU participants. When included in a model of prevalent CAD, 48 scores had practically equivalent Brier scores and AUROCs (region of practical equivalence = 0.02). Across these scores, 84% of participants had at least one score in both the top and bottom risk quintile. Continuous agreement of individual risk predictions from the 48 scores was poor, with an ICC of 0.351 (95% CI; 0.349, 0.352). Agreement between two statistically equivalent scores was moderate, with an ICC of 0.649 (95% CI; 0.646, 0.652). Light's Kappa, used to evaluate consistency of assignment to high-risk thresholds, did not exceed 0.56 (interpreted as 'fair') across statistically and practically equivalent scores. Repeating the analysis among 41,193 PMBB and 50,748 UCLA participants yielded different sets of statistically and practically equivalent scores which also lacked strong individual agreement. Conclusions and Relevance Across three diverse biobanks, CAD PRSs that performed equivalently at the population level produced unreliable individual risk estimates. Approaches to clinical implementation of CAD PRSs must consider the potential for discordant individual risk estimates from otherwise indistinguishable scores.
Collapse
|
5
|
Lee S, Shin D. A combination of red and processed meat intake and polygenic risk score influences the incidence of hyperuricemia in middle-aged Korean adults. Nutr Res Pract 2024; 18:721-745. [PMID: 39398885 PMCID: PMC11464275 DOI: 10.4162/nrp.2024.18.5.721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/03/2024] [Accepted: 08/22/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND/OBJECTIVES The high consumption of purine-rich meat is associated with hyperuricemia. However, there is limited evidence linking the consumption of red and processed meat to the genetic risk of hyperuricemia. We investigated the relationship between various combinations of red and processed meat consumption and the polygenic risk scores (PRSs) and the incidence of hyperuricemia in middle-aged Koreans. SUBJECTS/METHODS We analyzed the data from 44,053 participants aged ≥40 years sourced from the Health Examinees (HEXA) cohort of the Korean Genome and Epidemiology Study (KoGES). Information regarding red and processed meat intake was obtained using a semiquantitative food frequency questionnaire (SQ-FFQ). We identified 69 independent single-nucleotide polymorphisms (SNPs) at uric acid-related loci using genome-wide association studies (GWASs) and clumping analyses. The individual PRS, which is the weighted sum of the effect size of each allele at the SNP, was calculated. We used multivariable Cox proportional hazards models adjusted for covariates to determine the relationship between red and processed meat intake and the PRS in the incidence of hyperuricemia. RESULTS During an average follow-up period of 5 years, 2,556 patients with hyperuricemia were identified. For both men and women, the group with the highest red and processed meat intake and the highest PRS was positively associated with the development of hyperuricemia when compared with the group with the lowest red and processed meat intake and the lowest PRS (hazard ratio [HR], 2.72; 95% confidence interval [CI], 2.10-3.53; P < 0.0001; HR, 3.28; 95% CI, 2.45-4.40; P < 0.0001). CONCLUSION Individuals at a high genetic risk for uric acid levels should moderate their consumption of red and processed meat to prevent hyperuricemia.
Collapse
Affiliation(s)
- Suyeon Lee
- Department of Food and Nutrition, Inha University, Incheon 22212, Korea
| | - Dayeon Shin
- Department of Food and Nutrition, Inha University, Incheon 22212, Korea
| |
Collapse
|
6
|
Shaked O, Loza BL, Olthoff KM, Reddy KR, Keating BJ, Testa G, Asrani SK, Shaked A. Donor and recipient genetics: Implications for the development of posttransplant diabetes mellitus. Am J Transplant 2024; 24:1794-1802. [PMID: 38782187 DOI: 10.1016/j.ajt.2024.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 04/16/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024]
Abstract
Posttransplant diabetes mellitus (PTDM) is a prevalent complication of liver transplantation and is associated with cardiometabolic complications. We studied the consequences of genetic effects of liver donors and recipients on PTDM outcomes, focusing on the diverse genetic pathways related to insulin that play a role in the development of PTDM. One thousand one hundred fifteen liver transplant recipients without a pretransplant diagnosis of type 2 diabetes mellitus (T2D) and their paired donors recruited from 2 transplant centers had polygenic risk scores (PRS) for T2D, insulin secretion, and insulin sensitivity calculated. Among recipients in the highest T2D-PRS quintile, donor T2D-PRS did not contribute significantly to PTDM. However, in recipients with the lowest T2D genetic risk, donor livers with the highest T2D-PRS contributed to the development of PTDM (OR [95% CI] = 3.79 [1.10-13.1], P = .035). Recipient risk was linked to factors associated with insulin secretion (OR [95% CI] = 0.85 [0.74-0.98], P = .02), while donor livers contributed to PTDM via gene pathways involved in insulin sensitivity (OR [95% CI] = 0.86 [0.75-0.99], P = .03). Recipient and donor PRS independently and collectively serve as predictors of PTDM onset. The genetically influenced biological pathways in recipients primarily pertain to insulin secretion, whereas the genetic makeup of donors exerts an influence on insulin sensitivity.
Collapse
Affiliation(s)
- Oren Shaked
- Department of Surgery, Penn Transplant Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Bao-Li Loza
- Department of Surgery, Penn Transplant Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kim M Olthoff
- Department of Surgery, Penn Transplant Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kuchikula Rajender Reddy
- Department of Surgery, Penn Transplant Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Brendan J Keating
- Department of Surgery, Penn Transplant Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Giuliano Testa
- Annette C. and Harold C. Simmons Transplant Institute, Baylor University Medical Center, Dallas, TX, USA
| | - Sumeet K Asrani
- Annette C. and Harold C. Simmons Transplant Institute, Baylor University Medical Center, Dallas, TX, USA
| | - Abraham Shaked
- Department of Surgery, Penn Transplant Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
7
|
Shah S. Genomics for Improving Heart Failure Risk Assessment in Cancer Patients. JACC CardioOncol 2024; 6:728-730. [PMID: 39479318 PMCID: PMC11520204 DOI: 10.1016/j.jaccao.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2024] Open
Affiliation(s)
- Sonia Shah
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
| |
Collapse
|
8
|
Capalbo A, de Wert G, Mertes H, Klausner L, Coonen E, Spinella F, Van de Velde H, Viville S, Sermon K, Vermeulen N, Lencz T, Carmi S. Screening embryos for polygenic disease risk: a review of epidemiological, clinical, and ethical considerations. Hum Reprod Update 2024; 30:529-557. [PMID: 38805697 PMCID: PMC11369226 DOI: 10.1093/humupd/dmae012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/25/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND The genetic composition of embryos generated by in vitro fertilization (IVF) can be examined with preimplantation genetic testing (PGT). Until recently, PGT was limited to detecting single-gene, high-risk pathogenic variants, large structural variants, and aneuploidy. Recent advances have made genome-wide genotyping of IVF embryos feasible and affordable, raising the possibility of screening embryos for their risk of polygenic diseases such as breast cancer, hypertension, diabetes, or schizophrenia. Despite a heated debate around this new technology, called polygenic embryo screening (PES; also PGT-P), it is already available to IVF patients in some countries. Several articles have studied epidemiological, clinical, and ethical perspectives on PES; however, a comprehensive, principled review of this emerging field is missing. OBJECTIVE AND RATIONALE This review has four main goals. First, given the interdisciplinary nature of PES studies, we aim to provide a self-contained educational background about PES to reproductive specialists interested in the subject. Second, we provide a comprehensive and critical review of arguments for and against the introduction of PES, crystallizing and prioritizing the key issues. We also cover the attitudes of IVF patients, clinicians, and the public towards PES. Third, we distinguish between possible future groups of PES patients, highlighting the benefits and harms pertaining to each group. Finally, our review, which is supported by ESHRE, is intended to aid healthcare professionals and policymakers in decision-making regarding whether to introduce PES in the clinic, and if so, how, and to whom. SEARCH METHODS We searched for PubMed-indexed articles published between 1/1/2003 and 1/3/2024 using the terms 'polygenic embryo screening', 'polygenic preimplantation', and 'PGT-P'. We limited the review to primary research papers in English whose main focus was PES for medical conditions. We also included papers that did not appear in the search but were deemed relevant. OUTCOMES The main theoretical benefit of PES is a reduction in lifetime polygenic disease risk for children born after screening. The magnitude of the risk reduction has been predicted based on statistical modelling, simulations, and sibling pair analyses. Results based on all methods suggest that under the best-case scenario, large relative risk reductions are possible for one or more diseases. However, as these models abstract several practical limitations, the realized benefits may be smaller, particularly due to a limited number of embryos and unclear future accuracy of the risk estimates. PES may negatively impact patients and their future children, as well as society. The main personal harms are an unindicated IVF treatment, a possible reduction in IVF success rates, and patient confusion, incomplete counselling, and choice overload. The main possible societal harms include discarded embryos, an increasing demand for 'designer babies', overemphasis of the genetic determinants of disease, unequal access, and lower utility in people of non-European ancestries. Benefits and harms will vary across the main potential patient groups, comprising patients already requiring IVF, fertile people with a history of a severe polygenic disease, and fertile healthy people. In the United States, the attitudes of IVF patients and the public towards PES seem positive, while healthcare professionals are cautious, sceptical about clinical utility, and concerned about patient counselling. WIDER IMPLICATIONS The theoretical potential of PES to reduce risk across multiple polygenic diseases requires further research into its benefits and harms. Given the large number of practical limitations and possible harms, particularly unnecessary IVF treatments and discarded viable embryos, PES should be offered only within a research context before further clarity is achieved regarding its balance of benefits and harms. The gap in attitudes between healthcare professionals and the public needs to be narrowed by expanding public and patient education and providing resources for informative and unbiased genetic counselling.
Collapse
Affiliation(s)
- Antonio Capalbo
- Juno Genetics, Department of Reproductive Genetics, Rome, Italy
- Center for Advanced Studies and Technology (CAST), Department of Medical Genetics, “G. d’Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Guido de Wert
- Department of Health, Ethics & Society, CAPHRI-School for Public Health and Primary Care and GROW School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Heidi Mertes
- Department of Philosophy and Moral Sciences, Ghent University, Ghent, Belgium
- Department of Public Health and Primary Care, Ghent University, Ghent, Belgium
| | - Liraz Klausner
- Braun School of Public Health and Community Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Edith Coonen
- Departments of Clinical Genetics and Reproductive Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands
- School for Oncology and Developmental Biology, GROW, Maastricht University, Maastricht, The Netherlands
| | - Francesca Spinella
- Eurofins GENOMA Group Srl, Molecular Genetics Laboratories, Department of Scientific Communication, Rome, Italy
| | - Hilde Van de Velde
- Research Group Genetics Reproduction and Development (GRAD), Vrije Universiteit Brussel, Brussel, Belgium
- Brussels IVF, UZ Brussel, Brussel, Belgium
| | - Stephane Viville
- Laboratoire de Génétique Médicale LGM, Institut de Génétique Médicale d’Alsace IGMA, INSERM UMR 1112, Université de Strasbourg, France
- Laboratoire de Diagnostic Génétique, Unité de Génétique de l’infertilité (UF3472), Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Karen Sermon
- Research Group Genetics Reproduction and Development (GRAD), Vrije Universiteit Brussel, Brussel, Belgium
| | | | - Todd Lencz
- Institute of Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Departments of Psychiatry and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Shai Carmi
- Braun School of Public Health and Community Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
9
|
Sherafati A, Norland K, Naderian M, Schaid DJ, Kullo IJ. Polygenic Risk and Coronary Artery Disease Severity. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2024; 17:e004470. [PMID: 39114909 DOI: 10.1161/circgen.123.004470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 06/16/2024] [Indexed: 10/17/2024]
Abstract
BACKGROUND Coronary atherosclerotic burden and adverse coronary heart disease events are related phenotypes with likely shared genetic cause. METHODS We analyzed 6021 patients with available coronary angiography, genotyping, and exome sequencing data. We tested for associations of polygenic risk scores for coronary heart disease (PRSCHD) with multiple measures of coronary artery disease (CAD) severity. We assessed the joint associations of PRSCHD and pathogenic/likely pathogenic variants in 3 familial hypercholesterolemia genes, with CAD severity. We performed mediation analyses to explore whether CAD severity mediated the association of PRSCHD with prevalent coronary heart disease and incident myocardial infarction. RESULTS A 1-SD increase in PRSCHD was associated with multiple measures of CAD severity, including the log Gensini score (β, 0.31 [95% CI, 0.28-0.33]). Carrying a pathogenic/likely pathogenic familial hypercholesterolemia variant was associated with a higher log Gensini score after adjustment for PRSCHD (β, 0.21 [95% CI, 0.03-0.38]). A 1-SD increase in PRSCHD was associated with incident myocardial infarction over a mean follow-up of 9.2 years (hazard ratio, 1.20 [95% CI, 1.13-1.27]; P=5×10-10), and the Gensini score mediated 90% of this association. CONCLUSIONS PRSCHD was associated with multiple measures of CAD severity. The association of PRSCHD with incident myocardial infarction was almost fully mediated by CAD severity, indicating a considerable genetic overlap between the 2 phenotypes.
Collapse
Affiliation(s)
- Alborz Sherafati
- Department of Cardiovascular Medicine (A.S., K.N., M.N., I.J.K.)
| | - Kristjan Norland
- Department of Cardiovascular Medicine (A.S., K.N., M.N., I.J.K.)
| | | | | | - Iftikhar J Kullo
- Department of Cardiovascular Medicine (A.S., K.N., M.N., I.J.K.)
- Gonda Vascular Center, Mayo Clinic, Rochester, MN (I.J.K.)
| |
Collapse
|
10
|
Hutten CG, Boehm FJ, Smith JA, Spitzer BW, Wassertheil-Smoller S, Isasi CR, Cai J, Unkart JT, Sun J, Persky V, Daviglus ML, Sofer T, Argos M. Differential prediction performance between Caribbean- and Mainland-subgroups using state-of-the-art polygenic risk scores for coronary heart disease: Findings from the Hispanic Community Health Study/Study of Latinos (HCHS/SOL). MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.25.24313663. [PMID: 39399039 PMCID: PMC11469406 DOI: 10.1101/2024.09.25.24313663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Background Coronary heart disease (CHD) is a leading cause of death for Hispanic/Latino populations in the United States. We evaluated polygenic risk scores (PRS) with incident myocardial infarction (MI) in a Hispanic/Latino study sample. Methods We leveraged data from the Hispanic Community Health Study/Study of Latinos (HCHS/SOL) to assess four CHD-PRS from the PGS catalog, derived using multiple methods (LDpred, AnnoPred, stacked clumping and thresholding, and LDPred2). We evaluated associations between each standardized PRS and time to adjudicated incident MI, adjusted for age, sex, first 5 principal components, and weighted for survey design. Concordance statistics (c-index) compared predictive accuracy of each PRS with, and in addition to, traditional risk factors (TRF) for CHD (obesity, hypercholesterolemia, hypertension, diabetes, and smoking). Analyses were stratified by self-reported Caribbean- (Puerto Rican, Dominican or Cuban) and Mainland-(those of Mexican, Central American, or South American) heritage subgroups. Results After 11 years follow-up, for 9055 participants (mean age (SD) 47.6(13.1), 62.2% female), the incidence of MI was 1.0% (n = 95). Each PRS was more strongly associated with MI among Mainland participants. LDPred2 + TRF performed best among the Mainland subgroup; HR=2.69, 95% CI [1.71, 4.20], c-index = 0.897, 95% CI [0.848, 0.946]; a modest increase over TRF alone, c-index = 0.880, 95% CI [0.827, 0.933]. AnnoPred + TRF performed best among the Caribbean sample; c-index = 0.721, 95% CI [0.647, 0.795]; however, was not significantly associated with rate of MI (HR=1.14, 95% CI [0.82, 1.60]). Conclusion PRS performance for CHD is lacking for Hispanics/Latinos of Caribbean origin who have substantial proportions of African genetic ancestry, risking increased health disparities. AnnoPred, using functional annotations, outperformed other PRS in the Caribbean subgroup, suggesting a potential strategy for PRS construction in diverse populations. These results underscore the need to optimize cumulative genetic risk prediction of CHD in diverse Hispanic/Latino populations.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Maria Argos
- University of Illinois Chicago; Boston University
| |
Collapse
|
11
|
Debernardi C, Savoca A, De Gregorio A, Casalone E, Rosselli M, Herman EJ, Di Primio C, Tumino R, Sieri S, Vineis P, Panico S, Sacerdote C, Ardissino D, Asselta R, Matullo G. Population Heterogeneity and Selection of Coronary Artery Disease Polygenic Scores. J Pers Med 2024; 14:1025. [PMID: 39452533 PMCID: PMC11508882 DOI: 10.3390/jpm14101025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/16/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND/OBJECTIVES The identification of coronary artery disease (CAD) high-risk individuals is a major clinical need for timely diagnosis and intervention. Many different polygenic scores (PGSs) for CAD risk are available today to estimate the genetic risk. It is necessary to carefully choose the score to use, in particular for studies on populations, which are not adequately represented in the large datasets of European biobanks, such as the Italian one. This work aimed to analyze which PGS had the best performance within the Italian population. METHODS We used two Italian independent cohorts: the EPICOR case-control study (576 individuals) and the Atherosclerosis, Thrombosis, and Vascular Biology (ATVB) Italian study (3359 individuals). We evaluated 266 PGS for cardiovascular disease risk from the PGS Catalog, selecting 51 for CAD. RESULTS Distributions between patients and controls were significantly different for 49 scores (p-value < 0.01). Only five PGS have been trained and tested for the European population specifically. PGS003727 demonstrated to be the most accurate when evaluated independently (EPICOR AUC = 0.68; ATVB AUC = 0.80). Taking into account the conventional CAD risk factors further enhanced the performance of the model, particularly in the ATVB study (p-value = 0.0003). CONCLUSIONS European CAD PGS could have different risk estimates in peculiar populations, such as the Italian one, as well as in various geographical macro areas. Therefore, further evaluation is recommended for clinical applicability.
Collapse
Affiliation(s)
- Carla Debernardi
- Genomic Variation, Complex Diseases and Population Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (C.D.); (A.S.); (A.D.G.); (E.C.); (M.R.); (E.J.H.); (C.D.P.)
| | - Angelo Savoca
- Genomic Variation, Complex Diseases and Population Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (C.D.); (A.S.); (A.D.G.); (E.C.); (M.R.); (E.J.H.); (C.D.P.)
| | - Alessandro De Gregorio
- Genomic Variation, Complex Diseases and Population Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (C.D.); (A.S.); (A.D.G.); (E.C.); (M.R.); (E.J.H.); (C.D.P.)
| | - Elisabetta Casalone
- Genomic Variation, Complex Diseases and Population Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (C.D.); (A.S.); (A.D.G.); (E.C.); (M.R.); (E.J.H.); (C.D.P.)
| | - Miriam Rosselli
- Genomic Variation, Complex Diseases and Population Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (C.D.); (A.S.); (A.D.G.); (E.C.); (M.R.); (E.J.H.); (C.D.P.)
| | - Elton Jalis Herman
- Genomic Variation, Complex Diseases and Population Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (C.D.); (A.S.); (A.D.G.); (E.C.); (M.R.); (E.J.H.); (C.D.P.)
| | - Cecilia Di Primio
- Genomic Variation, Complex Diseases and Population Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (C.D.); (A.S.); (A.D.G.); (E.C.); (M.R.); (E.J.H.); (C.D.P.)
| | - Rosario Tumino
- Cancer Registry and Histopathology Unit, Azienda Ospedaliera “Civile-M.P. Arezzo”, 97100 Ragusa, Italy;
| | - Sabina Sieri
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, 20100 Milan, Italy;
| | - Paolo Vineis
- MRC-PHE Centre for Environment and Health, Imperial College London, London W12 0BZ, UK;
| | - Salvatore Panico
- Department of Clinical and Experimental Medicine, University Federico II, 80100 Naples, Italy;
| | - Carlotta Sacerdote
- Piedmont Reference Centre for Epidemiology and Cancer Prevention (CPO Piemonte), 10126 Turin, Italy;
| | - Diego Ardissino
- Cardiology Department, Azienda Ospedaliero-Universitaria of Parma, 43100 Parma, Italy;
- Department of Medicine and Surgery, University of Parma, 43100 Parma, Italy
| | - Rosanna Asselta
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy;
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Giuseppe Matullo
- Genomic Variation, Complex Diseases and Population Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (C.D.); (A.S.); (A.D.G.); (E.C.); (M.R.); (E.J.H.); (C.D.P.)
- Medical Genetic Service, Città della Salute e della Scienza, 10126 Turin, Italy
| |
Collapse
|
12
|
Wu T, Ke Y, Li Y, Wu Z, Lv J, Yu C, Sun D, Yao P, Kartsonaki C, Chen Z, Li L, Pang Y. Associations between circulating proteins and cardiometabolic diseases: a systematic review and meta-analysis of observational and Mendelian randomisation studies. Heart 2024; 110:1208-1215. [PMID: 39084708 DOI: 10.1136/heartjnl-2024-324050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 07/09/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Integration of large proteomics and genetic data in population-based studies can provide insights into discovery of novel biomarkers and potential therapeutic targets for cardiometabolic diseases (CMD). We aimed to synthesise existing evidence on the observational and genetic associations between circulating proteins and CMD. METHODS PubMed, Embase and Web of Science were searched until July 2023 for potentially relevant prospective observational and Mendelian randomisation (MR) studies investigating associations between circulating proteins and CMD, including coronary heart disease, stroke, type 2 diabetes, heart failure, atrial fibrillation and atherosclerosis. Two investigators independently extracted study characteristics using a standard form and pooled data using random effects models. RESULTS 50 observational, 25 MR and 10 studies performing both analyses were included, involving 26 414 160 non-overlapping participants. Meta-analysis of observational studies revealed 560 proteins associated with CMD, of which 133 proteins were associated with ≥2 CMDs (ie, pleiotropic). There were 245 potentially causal protein biomarkers identified in MR pooled results, involving 23 pleiotropic proteins. IL6RA and MMP12 were each causally associated with seven diseases. 22 protein-disease pairs showed directionally concordant associations in observational and MR pooled estimates. Addition of protein biomarkers to traditional clinical models modestly improved the accuracy of predicting incident CMD, with the highest improvement for heart failure (ΔC-index ~0.2). Of the 245 potentially causal proteins (291 protein-disease pairs), 3 pairs were validated by evidence of drug development from existing drug databases, 288 pairs lacked evidence of drug development and 66 proteins were drug targets approved for other indications. CONCLUSIONS Combined analyses of observational and genetic studies revealed the potential causal role of several proteins in the aetiology of CMD. Novel protein biomarkers are promising targets for drug development and risk stratification. PROSPERO REGISTRATION NUMBER CRD42022350327.
Collapse
Affiliation(s)
- Ting Wu
- School of Public Health, Peking University, Beijing, China
| | - Yalei Ke
- School of Public Health, Peking University, Beijing, China
| | - Yingtao Li
- School of Public Health, Peking University, Beijing, China
| | - Zhiyu Wu
- School of Public Health, Peking University, Beijing, China
| | - Jun Lv
- School of Public Health, Peking University, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education of the People's Republic of China, Beijing, China
| | - Canqing Yu
- School of Public Health, Peking University, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education of the People's Republic of China, Beijing, China
| | - Dianjianyi Sun
- School of Public Health, Peking University, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education of the People's Republic of China, Beijing, China
| | - Pang Yao
- Medical Research Council Population Health Research Unit at the University of Oxford, Oxford, UK
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Christiana Kartsonaki
- Medical Research Council Population Health Research Unit at the University of Oxford, Oxford, UK
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Zhengming Chen
- Medical Research Council Population Health Research Unit at the University of Oxford, Oxford, UK
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Liming Li
- School of Public Health, Peking University, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education of the People's Republic of China, Beijing, China
| | - Yuanjie Pang
- School of Public Health, Peking University, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education of the People's Republic of China, Beijing, China
| |
Collapse
|
13
|
Mehta UM, Roy N, Bahuguna A, Kotambail A, Arunachal G, Venkatasubramanian G, Thirthalli J. Incremental predictive value of genetic risk and functional brain connectivity in determining antipsychotic response in schizophrenia. Psychiatry Res 2024; 342:116201. [PMID: 39321637 DOI: 10.1016/j.psychres.2024.116201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 09/11/2024] [Accepted: 09/15/2024] [Indexed: 09/27/2024]
Abstract
We aimed to assess the incremental value of schizophrenia polygenic risk score (PgRS) and resting-state functional brain connectivity (rsFC) when added to clinical data in predicting the six-week response to oral risperidone (Risperdal) in schizophrenia. Fifty-seven, 54, and 43 individuals in a group of never-before-treated first-episode schizophrenia had good quality whole-genome sequencing (10x), rsFC, and both genomic and rsFC data, respectively, at baseline. Symptom severity ratings were obtained at baseline and six-weeks of oral risperidone (Risperdal) treatment. The primary outcome was the percentage change in the Positive and Negative Syndrome Scale Total scores after risperidone (Risperdal) treatment. Clinical, PgRS, and rsFC determinants of treatment response were first evaluated independently. Subsequently, three blocks of hierarchical multiple regression analyses with leave-one-out cross-validation (n = 43), were implemented to study clinical, clinical + PgRS and clinical + PgRS + rsFC determinants of treatment response. While the combined clinical variables did not show a statistically significant prediction of treatment response, adding PgRS (9 % R2 change) and rsFC (26 % R2 change) in hierarchical steps, significantly improved the overall proportion of variance explained in treatment response. This proof-of-concept investigation underscores the incremental benefits offered by genetic and neuroimaging metrics over clinical measures in determining prospectively-ascertained short-term treatment response in first-episode schizophrenia.
Collapse
Affiliation(s)
- Urvakhsh Meherwan Mehta
- Department of Psychiatry, National Institute of Mental Health and Neuro-Sciences (NIMHANS), Bangalore 560029, India.
| | - Neelabja Roy
- Department of Psychiatry, National Institute of Mental Health and Neuro-Sciences (NIMHANS), Bangalore 560029, India
| | - Ashutosh Bahuguna
- Department of Psychiatry, National Institute of Mental Health and Neuro-Sciences (NIMHANS), Bangalore 560029, India
| | - Ananthapadmanabha Kotambail
- Department of Human Genetics, National Institute of Mental Health and Neuro-Sciences (NIMHANS), Bangalore 560029, India
| | - Gautham Arunachal
- Department of Human Genetics, National Institute of Mental Health and Neuro-Sciences (NIMHANS), Bangalore 560029, India
| | - Ganesan Venkatasubramanian
- Department of Psychiatry, National Institute of Mental Health and Neuro-Sciences (NIMHANS), Bangalore 560029, India
| | - Jagadisha Thirthalli
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India
| |
Collapse
|
14
|
He Y, Lu W, Jee YH, Wang Y, Tsuo K, Qian DC, Diao JA, Huang H, Patel CJ, Byun J, Pasaniuc B, Atkinson EG, Amos CI, Moll M, Cho MH, Martin AR. Multi-trait and multi-ancestry genetic analysis of comorbid lung diseases and traits improves genetic discovery and polygenic risk prediction. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.25.24312558. [PMID: 39252935 PMCID: PMC11383478 DOI: 10.1101/2024.08.25.24312558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
While respiratory diseases such as COPD and asthma share many risk factors, most studies investigate them in insolation and in predominantly European ancestry populations. Here, we conducted the most powerful multi-trait and -ancestry genetic analysis of respiratory diseases and auxiliary traits to date. Our approach improves the power of genetic discovery across traits and ancestries, identifying 44 novel loci associated with lung function in individuals of East Asian ancestry. Using these results, we developed PRSxtra (cross TRait and Ancestry), a multi-trait and -ancestry polygenic risk score approach that leverages shared components of heritable risk via pleiotropic effects. PRSxtra significantly improved the prediction of asthma, COPD, and lung cancer compared to trait- and ancestry-matched PRS in a multi-ancestry cohort from the All of Us Research Program, especially in diverse populations. PRSxtra identified individuals in the top decile with over four-fold odds of asthma and COPD compared to the first decile. Our results present a new framework for multi-trait and -ancestry studies of respiratory diseases to improve genetic discovery and polygenic prediction.
Collapse
Affiliation(s)
- Yixuan He
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Wenhan Lu
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yon Ho Jee
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Ying Wang
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Kristin Tsuo
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA, USA
| | - David C Qian
- Department of Thoracic Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James A Diao
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Hailiang Huang
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Chirag J Patel
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Jinyoung Byun
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Bogdan Pasaniuc
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Department of Computational Medicine, University of California, Los Angeles
| | - Elizabeth G Atkinson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Christopher I Amos
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Matthew Moll
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Section on Pulmonary, Critical Care, Sleep and Allergy, Department of Veteran Affairs, Boston Healthcare System, West Roxbury, MA, USA
| | - Michael H Cho
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Alicia R Martin
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
15
|
Samani NJ, Beeston E, Greengrass C, Riveros-McKay F, Debiec R, Lawday D, Wang Q, Budgeon CA, Braund PS, Bramley R, Kharodia S, Newton M, Marshall A, Krzeminski A, Zafar A, Chahal A, Heer A, Khunti K, Joshi N, Lakhani M, Farooqi A, Plagnol V, Donnelly P, Weale ME, Nelson CP. Polygenic risk score adds to a clinical risk score in the prediction of cardiovascular disease in a clinical setting. Eur Heart J 2024; 45:3152-3160. [PMID: 38848106 PMCID: PMC11379490 DOI: 10.1093/eurheartj/ehae342] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 04/10/2024] [Accepted: 05/16/2024] [Indexed: 09/08/2024] Open
Abstract
BACKGROUND AND AIMS A cardiovascular disease polygenic risk score (CVD-PRS) can stratify individuals into different categories of cardiovascular risk, but whether the addition of a CVD-PRS to clinical risk scores improves the identification of individuals at increased risk in a real-world clinical setting is unknown. METHODS The Genetics and the Vascular Health Check Study (GENVASC) was embedded within the UK National Health Service Health Check (NHSHC) programme which invites individuals between 40-74 years of age without known CVD to attend an assessment in a UK general practice where CVD risk factors are measured and a CVD risk score (QRISK2) is calculated. Between 2012-2020, 44,141 individuals (55.7% females, 15.8% non-white) who attended an NHSHC in 147 participating practices across two counties in England were recruited and followed. When 195 individuals (cases) had suffered a major CVD event (CVD death, myocardial infarction or acute coronary syndrome, coronary revascularisation, stroke), 396 propensity-matched controls with a similar risk profile were identified, and a nested case-control genetic study undertaken to see if the addition of a CVD-PRS to QRISK2 in the form of an integrated risk tool (IRT) combined with QRISK2 would have identified more individuals at the time of their NHSHC as at high risk (QRISK2 10-year CVD risk of ≥10%), compared with QRISK2 alone. RESULTS The distribution of the standardised CVD-PRS was significantly different in cases compared with controls (cases mean score .32; controls, -.18, P = 8.28×10-9). QRISK2 identified 61.5% (95% confidence interval [CI]: 54.3%-68.4%) of individuals who subsequently developed a major CVD event as being at high risk at their NHSHC, while the combination of QRISK2 and IRT identified 68.7% (95% CI: 61.7%-75.2%), a relative increase of 11.7% (P = 1×10-4). The odds ratio (OR) of being up-classified was 2.41 (95% CI: 1.03-5.64, P = .031) for cases compared with controls. In individuals aged 40-54 years, QRISK2 identified 26.0% (95% CI: 16.5%-37.6%) of those who developed a major CVD event, while the combination of QRISK2 and IRT identified 38.4% (95% CI: 27.2%-50.5%), indicating a stronger relative increase of 47.7% in the younger age group (P = .001). The combination of QRISK2 and IRT increased the proportion of additional cases identified similarly in women as in men, and in non-white ethnicities compared with white ethnicity. The findings were similar when the CVD-PRS was added to the atherosclerotic cardiovascular disease pooled cohort equations (ASCVD-PCE) or SCORE2 clinical scores. CONCLUSIONS In a clinical setting, the addition of genetic information to clinical risk assessment significantly improved the identification of individuals who went on to have a major CVD event as being at high risk, especially among younger individuals. The findings provide important real-world evidence of the potential value of implementing a CVD-PRS into health systems.
Collapse
Affiliation(s)
- Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
| | - Emma Beeston
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
| | - Chris Greengrass
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
| | | | - Radoslaw Debiec
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
| | - Daniel Lawday
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
| | - Qingning Wang
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
| | - Charley A Budgeon
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
- School of Population and Global Health, University of Western Australia, Perth WA 6009, Australia
| | - Peter S Braund
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
| | - Richard Bramley
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
| | - Shireen Kharodia
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
| | - Michelle Newton
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
| | - Andrea Marshall
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
| | | | - Azhar Zafar
- Diabetes Research Centre, University of Leicester, Leicester General Hospital, Leicester LE5 4PW, UK
- Diabetes and Cardiovascular Medicine General Practice Alliance Federation Research and Training Academy, Northampton NN2 6AL, UK
| | - Anuj Chahal
- South Leicestershire Medical Group, Kibworth Beauchamp LE8 0LG, UK
| | | | - Kamlesh Khunti
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
- Diabetes Research Centre, University of Leicester, Leicester General Hospital, Leicester LE5 4PW, UK
| | - Nitin Joshi
- Willowbrook Medical Centre, Leicester LE5 2NL, UK
| | - Mayur Lakhani
- Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Azhar Farooqi
- Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Vincent Plagnol
- Genomics plc, King Charles House, Park End Street, Oxford OX1 1 JD, UK
| | - Peter Donnelly
- Genomics plc, King Charles House, Park End Street, Oxford OX1 1 JD, UK
| | - Michael E Weale
- Genomics plc, King Charles House, Park End Street, Oxford OX1 1 JD, UK
| | - Christopher P Nelson
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
| |
Collapse
|
16
|
Pillutla V, Aragam KG. Polygenic scores in real-world cardiovascular risk prediction: the path forward for assessing worth? Eur Heart J 2024; 45:3161-3163. [PMID: 39056297 DOI: 10.1093/eurheartj/ehae442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/28/2024] Open
Affiliation(s)
- Virimchi Pillutla
- Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 3.128, 185 Cambridge St, Boston, MA 02114, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Krishna G Aragam
- Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 3.128, 185 Cambridge St, Boston, MA 02114, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
17
|
Oza A. Why does heart disease affect so many young South Asians? Nature 2024; 633:272-274. [PMID: 39261610 DOI: 10.1038/d41586-024-02916-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
|
18
|
Stein R, Ferrari F, García-Giustiniani D. Polygenic Risk Scores: The Next Step for Improved Risk Stratification in Coronary Artery Disease? Arq Bras Cardiol 2024; 121:e20240252. [PMID: 39352188 PMCID: PMC11495647 DOI: 10.36660/abc.20240252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 07/18/2024] [Indexed: 10/03/2024] Open
Abstract
Despite significant advances in the management of coronary artery disease (CAD) and reductions in annual mortality rates in recent decades, this disease remains the leading cause of death worldwide. Consequently, there is an ongoing need for efforts to address this situation. Current clinical algorithms to identify at-risk patients are particularly inaccurate in moderate-risk individuals. For this reason, the need for ancillary tests has been suggested, including predictive genetic screening. As genetic studies rapidly expand and genomic data becomes more accessible, numerous genetic risk scores have been proposed to identify and evaluate an individual's susceptibility to developing diseases, including CAD. The field of genetics has indeed made substantial contributions to risk prediction, particularly in cases where children have parents with premature CAD, resulting in an increased risk of up to 75%. The polygenic risk scores (PRSs) have emerged as a potentially valuable tool for understanding and stratifying an individual's genetic risk. The PRS is calculated as a weighted sum of single-nucleotide variants present throughout the human genome, identifiable through genome-wide association studies, and associated with various cardiometabolic diseases. The use of PRSs holds promise, as it enables the development of personalized strategies for preventing or diagnosing specific pathologies early. Furthermore, it can complement existing clinical scores, increasing the accuracy of individual risk prediction. Consequently, the application of PRSs has the potential to impact the costs and adverse outcomes associated with CAD positively. This narrative review provides an overview of the role of PRSs in the context of CAD.
Collapse
Affiliation(s)
- Ricardo Stein
- Programa de Pós-Graduação em Cardiologia e Ciências CardiovascularesUniversidade Federal do Rio Grande do SulPorto AlegreRSBrasilPrograma de Pós-Graduação em Cardiologia e Ciências Cardiovasculares, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS – Brasil
- Departamento de Medicina InternaUniversidade Federal do Rio Grande do SulPorto AlegreRSBrasilDepartamento de Medicina Interna, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS – Brasil
| | - Filipe Ferrari
- Programa de Pós-Graduação em Cardiologia e Ciências CardiovascularesUniversidade Federal do Rio Grande do SulPorto AlegreRSBrasilPrograma de Pós-Graduação em Cardiologia e Ciências Cardiovasculares, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS – Brasil
| | - Diego García-Giustiniani
- Instituto de Investigación Biomédica de A CoruñaCoruñaGaliciaEspanhaInstituto de Investigación Biomédica de A Coruña (INIBIC), Coruña, Galicia – Espanha
| |
Collapse
|
19
|
Sun D, Ding Y, Yu C, Sun D, Pang Y, Pei P, Yang L, Millwood IY, Walters RG, Du H, Chen X, Schmidt D, Stevens R, Chen J, Chen Z, Li L, Lv J. Joint impact of polygenic risk score and lifestyles on early- and late-onset cardiovascular diseases. Nat Hum Behav 2024; 8:1810-1818. [PMID: 38987358 DOI: 10.1038/s41562-024-01923-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 06/10/2024] [Indexed: 07/12/2024]
Abstract
Understanding the interactions between genetic risk and lifestyles on different types and age onsets of cardiovascular disease (CVD) risk can help identify individuals for whom lifestyle changes would be beneficial. Here we developed three polygenic risk scores, called MetaPRSs, for coronary artery disease, ischaemic stroke and intracerebral haemorrhage by combining PRSs for CVD and CVD-related risk factors in 96,400 participants from the prospective China Kadoorie Biobank. Genetic and lifestyle risks were categorized by the disease-specific MetaPRSs and the number of unfavourable lifestyles. High genetic risk and unfavourable lifestyles were found to be more strongly associated with early than late onset of CVD outcomes in men and women. Change from unfavourable to favourable lifestyles resulted in 14.7-, 2.5- and 2.6-fold greater reductions in incidence rates of early-onset coronary artery disease and ischaemic stroke and late-onset coronary artery disease in high than low genetic risk group. Young adults at high genetic risk may have larger benefits in preventing CVD from lifestyle improvements.
Collapse
|
20
|
Fujii R, Hishida A, Nakatochi M, Okumiyama H, Takashima N, Tsuboi Y, Suzuki K, Ikezaki H, Shimanoe C, Kato Y, Tamura T, Ito H, Michihata N, Tanoue S, Suzuki S, Kuriki K, Kadota A, Watanabe T, Momozawa Y, Wakai K, Matsuo K. Polygenic risk score for blood pressure and lifestyle factors with overall and CVD mortality: a prospective cohort study in a Japanese population. Hypertens Res 2024; 47:2284-2294. [PMID: 38961281 DOI: 10.1038/s41440-024-01766-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 05/29/2024] [Accepted: 06/06/2024] [Indexed: 07/05/2024]
Abstract
Although previous polygenic risk score (PRS) studies for cardiovascular disease (CVD) focused on incidence, few studies addressed CVD mortality and quantified risks by environmental exposures in different genetic liability groups. This prospective study aimed to examine the associations of blood pressure PRS with all-cause and CVD mortality and to quantify the attributable risk by modifiable lifestyles across different PRS strata. 9,296 participants in the Japan Multi-Institutional Collaborative Cohort Study without hypertension at baseline were analyzed in this analysis. PRS for systolic blood pressure and diastolic blood pressure (PRSSBP and PRSDBP) were developed using publicly available Biobank Japan GWAS summary statistics. CVD-related mortality was defined by the International Classification of Diseases 10th version (I00-I99). Cox-proportional hazard model was used to examine associations of PRSs and lifestyle variables (smoking, drinking, and dietary sodium intake) with mortality. During a median 12.6-year follow-up period, we observed 273 all-cause and 41 CVD mortality cases. Compared to the middle PRS group (20-80th percentile), adjusted hazard ratios for CVD mortality at the top PRS group ( > 90th percentile) were 3.67 for PRSSBP and 2.92 for PRSDBP. Attributable risks of CVD mortality by modifiable lifestyles were higher in the high PRS group ( > 80th percentile) compared with the low PRS group (0-80th percentile). In summary, blood pressure PRS is associated with CVD mortality in the general Japanese population. Our study implies that integrating PRS with lifestyle could contribute to identify target populations for lifestyle intervention even though improvement of discriminatory ability by PRS alone is limited.
Collapse
Affiliation(s)
- Ryosuke Fujii
- Department of Preventive Medical Sciences, Fujita Health University School of Medical Sciences, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, 470-1192, Japan.
- Institute for Biomedicine, Eurac Research, Via A.Volta 21, Bolzano/Bozen, 39100, Italy.
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Asahi Hishida
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Masahiro Nakatochi
- Public Health Informatics Unit, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, 1-1-20 Daikominami, Higashi-ku, Nagoya, 461-8673, Japan
| | - Hiroshi Okumiyama
- Department of Preventive Medical Sciences, Fujita Health University School of Medical Sciences, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, 470-1192, Japan
| | - Naoyuki Takashima
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
- NCD Epidemiology Research Center, Shiga University of Medical Science, Tsukiwacho, Seta, Otsu, 520-2192, Japan
| | - Yoshiki Tsuboi
- Department of Preventive Medical Sciences, Fujita Health University School of Medical Sciences, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, 470-1192, Japan
| | - Koji Suzuki
- Department of Preventive Medical Sciences, Fujita Health University School of Medical Sciences, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, 470-1192, Japan
| | - Hiroaki Ikezaki
- Department of Comprehensive General Internal Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
- Department of General Internal Medicine, Kyushu University Hospital, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Chisato Shimanoe
- Department of Pharmacy, Saga University Hospital, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Yasufumi Kato
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Takashi Tamura
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Hidemi Ito
- Division of Cancer Information and Control, Aichi Cancer Center Research Institute, 1-1 Kanokoden, Chikusa-ku, Nagoya, 464-8681, Japan
- Division of Descriptive Cancer Epidemiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Nobuaki Michihata
- Cancer Prevention Center, Chiba Cancer Center Research Institute, 666-2 Nitona-cho, Chuo-ku, Chiba, 260-8717, Japan
| | - Shiroh Tanoue
- Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Sadao Suzuki
- Department of Public Health, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Kiyonori Kuriki
- Laboratory of Public Health, Division of Nutritional Sciences, School of Food and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| | - Aya Kadota
- NCD Epidemiology Research Center, Shiga University of Medical Science, Tsukiwacho, Seta, Otsu, 520-2192, Japan
- Faculty of Nursing Science, Tsuruga Nursing University, 78-2 Kizaki, Tsuruga, 914-0814, Japan
| | - Takeshi Watanabe
- Department of Preventive Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Yukihide Momozawa
- Laboratory for Genotyping Development, Center for Integrative Medical Sciences, RIKEN, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Kenji Wakai
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Keitaro Matsuo
- Division of Cancer Information and Control, Aichi Cancer Center Research Institute, 1-1 Kanokoden, Chikusa-ku, Nagoya, 464-8681, Japan
- Division of Descriptive Cancer Epidemiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| |
Collapse
|
21
|
Marston NA. Polygenic Risk and the Progression of High-Risk Coronary Plaque. JACC Cardiovasc Imaging 2024:S1936-878X(24)00316-4. [PMID: 39269416 DOI: 10.1016/j.jcmg.2024.07.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 07/24/2024] [Indexed: 09/15/2024]
Affiliation(s)
- Nicholas A Marston
- TIMI Study Group, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
22
|
Clarke SL, Huang RDL, Hilliard AT, Levin MG, Sharma D, Thomson B, Lynch J, Tsao PS, Gaziano JM, Assimes TL. Genetically predicted lipoprotein(a) associates with coronary artery plaque severity independent of low-density lipoprotein cholesterol. Eur J Prev Cardiol 2024:zwae271. [PMID: 39158116 DOI: 10.1093/eurjpc/zwae271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/11/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
AIMS Elevated Lipoprotein(a) [Lp(a)] is a causal risk factor for atherosclerotic cardiovascular disease, but the mechanisms of risk are debated. Studies have found inconsistent associations between Lp(a) and measurements of atherosclerosis. We aimed to assess the relationship between Lp(a), low-density lipoprotein cholesterol (LDL-C) and coronary artery plaque severity. METHODS The study population consisted of participants of the Million Veteran Program who have undergone an invasive angiogram. The primary exposure was genetically predicted Lp(a), estimated by a polygenic score. Genetically predicted LDL-C was also assessed for comparison. The primary outcome was coronary artery plaque severity, categorized as normal, non-obstructive disease, 1-vessel disease, 2-vessel disease, and 3-vessel or left main disease. RESULTS Among 18,927 adults of genetically inferred European ancestry and 4,039 adults of genetically inferred African ancestry, we observed consistent associations between genetically predicted Lp(a) and obstructive coronary plaque, with effect sizes trending upward for increasingly severe categories of disease. Associations were independent of risk factors, clinically measured LDL-C and genetically predicted LDL-C. However, we did not find strong or consistent evidence for an association between genetically predicted Lp(a) and risk for non-obstructive plaque. CONCLUSIONS Genetically predicted Lp(a) is positively associated with coronary plaque severity independent of LDL-C, consistent with Lp(a) promoting atherogenesis. However, the effects of Lp(a) may be greater for progression of plaque to obstructive disease than for the initial development of non-obstructive plaque. A limitation of this study is that Lp(a) was estimated using genetic markers and could not be directly assayed, nor could apo(a) isoform size.
Collapse
Affiliation(s)
- Shoa L Clarke
- VA Palo Alto Healthcare System, Palo Alto, CA, USA
- Department of Medicine, Stanford Prevention Research Center, Stanford University School of Medicine, Palo Alto, CA, USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Rose D L Huang
- VA Palo Alto Healthcare System, Palo Alto, CA, USA
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
| | | | - Michael G Levin
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Medicine, Division of Cardiovascular Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Disha Sharma
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Blake Thomson
- Stanford University School of Medicine, Stanford, CA, USA
| | - Julie Lynch
- VA Informatics and Computing Infrastructure, VA Salt Lake City Health Care System, Salt Lake City, UT, USA
| | | | - J Michael Gaziano
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
| | - Themistocles L Assimes
- VA Palo Alto Healthcare System, Palo Alto, CA, USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
23
|
Chen T, Zhang H, Mazumder R, Lin X. Fast and scalable ensemble learning method for versatile polygenic risk prediction. Proc Natl Acad Sci U S A 2024; 121:e2403210121. [PMID: 39110727 PMCID: PMC11331062 DOI: 10.1073/pnas.2403210121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 07/11/2024] [Indexed: 08/21/2024] Open
Abstract
Polygenic risk scores (PRS) enhance population risk stratification and advance personalized medicine, but existing methods face several limitations, encompassing issues related to computational burden, predictive accuracy, and adaptability to a wide range of genetic architectures. To address these issues, we propose Aggregated L0Learn using Summary-level data (ALL-Sum), a fast and scalable ensemble learning method for computing PRS using summary statistics from genome-wide association studies (GWAS). ALL-Sum leverages a L0L2 penalized regression and ensemble learning across tuning parameters to flexibly model traits with diverse genetic architectures. In extensive large-scale simulations across a wide range of polygenicity and GWAS sample sizes, ALL-Sum consistently outperformed popular alternative methods in terms of prediction accuracy, runtime, and memory usage by 10%, 20-fold, and threefold, respectively, and demonstrated robustness to diverse genetic architectures. We validated the performance of ALL-Sum in real data analysis of 11 complex traits using GWAS summary statistics from nine data sources, including the Global Lipids Genetics Consortium, Breast Cancer Association Consortium, and FinnGen Biobank, with validation in the UK Biobank. Our results show that on average, ALL-Sum obtained PRS with 25% higher accuracy on average, with 15 times faster computation and half the memory than the current state-of-the-art methods, and had robust performance across a wide range of traits and diseases. Furthermore, our method demonstrates stable prediction when using linkage disequilibrium computed from different data sources. ALL-Sum is available as a user-friendly R software package with publicly available reference data for streamlined analysis.
Collapse
Affiliation(s)
- Tony Chen
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA02215
| | - Haoyu Zhang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD20814
| | - Rahul Mazumder
- Operations Research and Statistics Group, Sloan School of Management, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Xihong Lin
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA02215
- Department of Statistics, Harvard University, Cambridge, MA02138
| |
Collapse
|
24
|
Sharew NT, Clark SR, Schubert KO, Amare AT. Pharmacogenomic scores in psychiatry: systematic review of current evidence. Transl Psychiatry 2024; 14:322. [PMID: 39107294 PMCID: PMC11303815 DOI: 10.1038/s41398-024-02998-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 06/21/2024] [Accepted: 06/27/2024] [Indexed: 08/10/2024] Open
Abstract
In the past two decades, significant progress has been made in the development of polygenic scores (PGSs). One specific application of PGSs is the development and potential use of pharmacogenomic- scores (PGx-scores) to identify patients who can benefit from a specific medication or are likely to experience side effects. This systematic review comprehensively evaluates published PGx-score studies in psychiatry and provides insights into their potential clinical use and avenues for future development. A systematic literature search was conducted across PubMed, EMBASE, and Web of Science databases until 22 August 2023. This review included fifty-three primary studies, of which the majority (69.8%) were conducted using samples of European ancestry. We found that over 90% of PGx-scores in psychiatry have been developed based on psychiatric and medical diagnoses or trait variants, rather than pharmacogenomic variants. Among these PGx-scores, the polygenic score for schizophrenia (PGSSCZ) has been most extensively studied in relation to its impact on treatment outcomes (32 publications). Twenty (62.5%) of these studies suggest that individuals with higher PGSSCZ have negative outcomes from psychotropic treatment - poorer treatment response, higher rates of treatment resistance, more antipsychotic-induced side effects, or more psychiatric hospitalizations, while the remaining studies did not find significant associations. Although PGx-scores alone accounted for at best 5.6% of the variance in treatment outcomes (in schizophrenia treatment resistance), together with clinical variables they explained up to 13.7% (in bipolar lithium response), suggesting that clinical translation might be achieved by including PGx-scores in multivariable models. In conclusion, our literature review found that there are still very few studies developing PGx-scores using pharmacogenomic variants. Research with larger and diverse populations is required to develop clinically relevant PGx-scores, using biology-informed and multi-phenotypic polygenic scoring approaches, as well as by integrating clinical variables with these scores to facilitate their translation to psychiatric practice.
Collapse
Affiliation(s)
- Nigussie T Sharew
- Discipline of Psychiatry, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Asrat Woldeyes Health Science Campus, Debre Berhan University, Debre Berhan, Ethiopia
| | - Scott R Clark
- Discipline of Psychiatry, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - K Oliver Schubert
- Discipline of Psychiatry, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Division of Mental Health, Northern Adelaide Local Health Network, SA Health, Adelaide, Australia
- Headspace Adelaide Early Psychosis - Sonder, Adelaide, SA, Australia
| | - Azmeraw T Amare
- Discipline of Psychiatry, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia.
| |
Collapse
|
25
|
Tsuo K, Shi Z, Ge T, Mandla R, Hou K, Ding Y, Pasaniuc B, Wang Y, Martin AR. All of Us diversity and scale improve polygenic prediction contextually with greatest improvements for under-represented populations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.06.606846. [PMID: 39149254 PMCID: PMC11326295 DOI: 10.1101/2024.08.06.606846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Recent studies have demonstrated that polygenic risk scores (PRS) trained on multi-ancestry data can improve prediction accuracy in groups historically underrepresented in genomic studies, but the availability of linked health and genetic data from large-scale diverse cohorts representative of a wide spectrum of human diversity remains limited. To address this need, the All of Us research program (AoU) generated whole-genome sequences of 245,388 individuals who collectively reflect the diversity of the USA. Leveraging this resource and another widely-used population-scale biobank, the UK Biobank (UKB) with a half million participants, we developed PRS trained on multi-ancestry and multi-biobank data with up to ~750,000 participants for 32 common, complex traits and diseases across a range of genetic architectures. We then compared effects of ancestry, PRS methodology, and genetic architecture on PRS accuracy across a held out subset of ancestrally diverse AoU participants. Due to the more heterogeneous study design of AoU, we found lower heritability on average compared to UKB (0.075 vs 0.165), which limited the maximal achievable PRS accuracy in AoU. Overall, we found that the increased diversity of AoU significantly improved PRS performance in some participants in AoU, especially underrepresented individuals, across multiple phenotypes. Notably, maximizing sample size by combining discovery data across AoU and UKB is not the optimal approach for predicting some phenotypes in African ancestry populations; rather, using data from only AoU for these traits resulted in the greatest accuracy. This was especially true for less polygenic traits with large ancestry-enriched effects, such as neutrophil count (R 2: 0.055 vs. 0.035 using AoU vs. cross-biobank meta-analysis, respectively, because of e.g. DARC). Lastly, we calculated individual-level PRS accuracies rather than grouping by continental ancestry, a critical step towards interpretability in precision medicine. Individualized PRS accuracy decays linearly as a function of ancestry divergence, but the slope was smaller using multi-ancestry GWAS compared to using European GWAS. Our results highlight the potential of biobanks with more balanced representations of human diversity to facilitate more accurate PRS for the individuals least represented in genomic studies.
Collapse
Affiliation(s)
- Kristin Tsuo
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Zhuozheng Shi
- Interdepartmental Program in Bioinformatics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Tian Ge
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Precision Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Ravi Mandla
- Interdepartmental Program in Bioinformatics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kangcheng Hou
- Interdepartmental Program in Bioinformatics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yi Ding
- Interdepartmental Program in Bioinformatics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Bogdan Pasaniuc
- Interdepartmental Program in Bioinformatics, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Institute for Precision Health, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Computational Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ying Wang
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alicia R Martin
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
26
|
Nurmohamed NS, Shim I, Gaillard EL, Ibrahim S, Bom MJ, Earls JP, Min JK, Planken RN, Choi AD, Natarajan P, Stroes ESG, Knaapen P, Reeskamp LF, Fahed AC. Polygenic Risk Is Associated With Long-Term Coronary Plaque Progression and High-Risk Plaque. JACC Cardiovasc Imaging 2024:S1936-878X(24)00253-5. [PMID: 39152960 DOI: 10.1016/j.jcmg.2024.06.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/24/2024] [Accepted: 06/28/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND The longitudinal relation between coronary artery disease (CAD) polygenic risk score (PRS) and long-term plaque progression and high-risk plaque (HRP) features is unknown. OBJECTIVES The goal of this study was to investigate the impact of CAD PRS on long-term coronary plaque progression and HRP. METHODS Patients underwent CAD PRS measurement and prospective serial coronary computed tomography angiography (CTA) imaging. Coronary CTA scans were analyzed with a previously validated artificial intelligence-based algorithm (atherosclerosis imaging-quantitative computed tomography imaging). The relationship between CAD PRS and change in percent atheroma volume (PAV), percent noncalcified plaque progression, and HRP prevalence was investigated in linear mixed-effect models adjusted for baseline plaque volume and conventional risk factors. RESULTS A total of 288 subjects (mean age 58 ± 7 years; 60% male) were included in this study with a median scan interval of 10.2 years. At baseline, patients with a high CAD PRS had a more than 5-fold higher PAV than those with a low CAD PRS (10.4% vs 1.9%; P < 0.001). Per 10 years of follow-up, a 1 SD increase in CAD PRS was associated with a 0.69% increase in PAV progression in the multivariable adjusted model. CAD PRS provided additional discriminatory benefit for above-median noncalcified plaque progression during follow-up when added to a model with conventional risk factors (AUC: 0.73 vs 0.69; P = 0.039). Patients with high CAD PRS had an OR of 2.85 (95% CI: 1.14-7.14; P = 0.026) and 6.16 (95% CI: 2.55-14.91; P < 0.001) for having HRP at baseline and follow-up compared with those with low CAD PRS. CONCLUSIONS Polygenic risk is strongly associated with future long-term plaque progression and HRP in patients suspected of having CAD.
Collapse
Affiliation(s)
- Nick S Nurmohamed
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Division of Cardiology, The George Washington University School of Medicine, Washington, DC, USA
| | - Injeong Shim
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA; Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Samsung Medical Center, Seoul, Republic of South Korea
| | - Emilie L Gaillard
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Shirin Ibrahim
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Michiel J Bom
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | | | | | - R Nils Planken
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Universiteit van Amsterdam, Amsterdam, the Netherlands
| | - Andrew D Choi
- Division of Cardiology, The George Washington University School of Medicine, Washington, DC, USA
| | - Pradeep Natarajan
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA; Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Erik S G Stroes
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Paul Knaapen
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Laurens F Reeskamp
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.
| | - Akl C Fahed
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA; Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
27
|
Judd J, Spence JP, Pritchard JK, Kachuri L, Witte JS. Investigating the Role of Neighborhood Socioeconomic Status and Germline Genetics on Prostate Cancer Risk. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.31.24311312. [PMID: 39132496 PMCID: PMC11312637 DOI: 10.1101/2024.07.31.24311312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Background Genetic factors play an important role in prostate cancer (PCa) development with polygenic risk scores (PRS) predicting disease risk across genetic ancestries. However, there are few convincing modifiable factors for PCa and little is known about their potential interaction with genetic risk. We analyzed incident PCa cases (n=6,155) and controls (n=98,257) of European and African ancestry from the UK Biobank (UKB) cohort to evaluate the role of neighborhood socioeconomic status (nSES)-and how it may interact with PRS-on PCa risk. Methods We evaluated a multi-ancestry PCa PRS containing 269 genetic variants to understand the association of germline genetics with PCa in UKB. Using the English Indices of Deprivation, a set of validated metrics that quantify lack of resources within geographical areas, we performed logistic regression to investigate the main effects and interactions between nSES deprivation, PCa PRS, and PCa. Results The PCa PRS was strongly associated with PCa (OR=2.04; 95%CI=2.00-2.09; P<0.001). Additionally, nSES deprivation indices were inversely associated with PCa: employment (OR=0.91; 95%CI=0.86-0.96; P<0.001), education (OR=0.94; 95%CI=0.83-0.98; P<0.001), health (OR=0.91; 95%CI=0.86-0.96; P<0.001), and income (OR=0.91; 95%CI=0.86-0.96; P<0.001). The PRS effects showed little heterogeneity across nSES deprivation indices, except for the Townsend Index (P=0.03). Conclusions We reaffirmed genetics as a risk factor for PCa and identified nSES deprivation domains that influence PCa detection and are potentially correlated with environmental exposures that are a risk factor for PCa. These findings also suggest that nSES and genetic risk factors for PCa act independently.
Collapse
Affiliation(s)
- Jonathan Judd
- Department of Genetics, Stanford University, Stanford CA
| | | | - Jonathan K. Pritchard
- Department of Genetics, Stanford University, Stanford CA
- Department of Biology, Stanford University, Stanford CA
| | - Linda Kachuri
- Department of Epidemiology and Population Health, Stanford University, Stanford CA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - John S. Witte
- Department of Genetics, Stanford University, Stanford CA
- Department of Epidemiology and Population Health, Stanford University, Stanford CA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
28
|
Singh S, Stocco G, Theken KN, Dickson A, Feng Q, Karnes JH, Mosley JD, El Rouby N. Pharmacogenomics polygenic risk score: Ready or not for prime time? Clin Transl Sci 2024; 17:e13893. [PMID: 39078255 PMCID: PMC11287822 DOI: 10.1111/cts.13893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 06/11/2024] [Accepted: 06/25/2024] [Indexed: 07/31/2024] Open
Abstract
Pharmacogenomic Polygenic Risk Scores (PRS) have emerged as a tool to address the polygenic nature of pharmacogenetic phenotypes, increasing the potential to predict drug response. Most pharmacogenomic PRS have been extrapolated from disease-associated variants identified by genome wide association studies (GWAS), although some have begun to utilize genetic variants from pharmacogenomic GWAS. As pharmacogenomic PRS hold the promise of enabling precision medicine, including stratified treatment approaches, it is important to assess the opportunities and challenges presented by the current data. This assessment will help determine how pharmacogenomic PRS can be advanced and transitioned into clinical use. In this review, we present a summary of recent evidence, evaluate the current status, and identify several challenges that have impeded the progress of pharmacogenomic PRS. These challenges include the reliance on extrapolations from disease genetics and limitations inherent to pharmacogenomics research such as low sample sizes, phenotyping inconsistencies, among others. We finally propose recommendations to overcome the challenges and facilitate the clinical implementation. These recommendations include standardizing methodologies for phenotyping, enhancing collaborative efforts, developing new statistical methods to capitalize on drug-specific genetic associations for PRS construction. Additional recommendations include enhancing the infrastructure that can integrate genomic data with clinical predictors, along with implementing user-friendly clinical decision tools, and patient education. Ethical and regulatory considerations should address issues related to patient privacy, informed consent and safe use of PRS. Despite these challenges, ongoing research and large-scale collaboration is likely to advance the field and realize the potential of pharmacogenomic PRS.
Collapse
Affiliation(s)
- Sonal Singh
- Merck & Co., IncSouth San FranciscoCaliforniaUSA
| | - Gabriele Stocco
- Department of Medical, Surgical and Health SciencesUniversity of TriesteTriesteItaly
- Institute for Maternal and Child Health IRCCS Burlo GarofoloTriesteItaly
| | - Katherine N. Theken
- Department of Oral and Maxillofacial Surgery and Pharmacology, School of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Alyson Dickson
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - QiPing Feng
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Jason H. Karnes
- Department of Pharmacy Practice and Science, R. Ken Coit College of PharmacyUniversity of ArizonaTucsonArizonaUSA
- Department of Biomedical InformaticsVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Jonathan D. Mosley
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
- Department of Biomedical InformaticsVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Nihal El Rouby
- Division of Pharmacy Practice and Adminstrative Sciences, James L Winkle College of PharmacyUniversity of CincinnatiCincinnatiOhioUSA
- St. Elizabeth HealthcareEdgewoodKentuckyUSA
| |
Collapse
|
29
|
Yu Z, Coorens THH, Uddin MM, Ardlie KG, Lennon N, Natarajan P. Genetic variation across and within individuals. Nat Rev Genet 2024; 25:548-562. [PMID: 38548833 PMCID: PMC11457401 DOI: 10.1038/s41576-024-00709-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2024] [Indexed: 04/12/2024]
Abstract
Germline variation and somatic mutation are intricately connected and together shape human traits and disease risks. Germline variants are present from conception, but they vary between individuals and accumulate over generations. By contrast, somatic mutations accumulate throughout life in a mosaic manner within an individual due to intrinsic and extrinsic sources of mutations and selection pressures acting on cells. Recent advancements, such as improved detection methods and increased resources for association studies, have drastically expanded our ability to investigate germline and somatic genetic variation and compare underlying mutational processes. A better understanding of the similarities and differences in the types, rates and patterns of germline and somatic variants, as well as their interplay, will help elucidate the mechanisms underlying their distinct yet interlinked roles in human health and biology.
Collapse
Affiliation(s)
- Zhi Yu
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | | | - Md Mesbah Uddin
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | | | - Niall Lennon
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Pradeep Natarajan
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
30
|
Kuś A, Sterenborg RBTM, Haug EB, Galesloot TE, Visser WE, Smit JWA, Bednarczuk T, Peeters RP, Åsvold BO, Teumer A, Medici M. Towards Personalized TSH Reference Ranges: A Genetic and Population-Based Approach in Three Independent Cohorts. Thyroid 2024; 34:969-979. [PMID: 38919119 DOI: 10.1089/thy.2024.0045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/27/2024]
Abstract
Background: Serum thyroid-stimulating hormone (TSH) measurement is the diagnostic cornerstone for primary thyroid dysfunction. There is high inter-individual but limited intra-individual variation in TSH concentrations, largely due to genetic factors. The currently used wide population-based reference intervals may lead to inappropriate management decisions. Methods: A polygenic score (PGS) including 59 genetic variants was used to calculate genetically determined TSH reference ranges in a thyroid disease-free cohort (n = 6,834). Its effect on reclassification of diagnoses was investigated when compared to using population-based reference ranges. Next, results were validated in a second independent population-based thyroid disease-free cohort (n = 3,800). Potential clinical implications were assessed in a third independent population-based cohort including individuals without thyroid disease (n = 26,321) as well as individuals on levothyroxine (LT4) treatment (n = 1,132). Results: PGS was a much stronger predictor of individual TSH concentrations than FT4 (total variance in TSH concentrations explained 9.2-11.1% vs. 2.4-2.7%, respectively) or any other nongenetic factor (total variance in TSH concentrations explained 0.2-1.8%). Genetically determined TSH reference ranges differed significantly between PGS quartiles in all cohorts, while the differences in FT4 concentrations were absent or only minor. Up to 24.7-30.1% of individuals, previously classified as having subclinical hypo- and hyperthyroidism when using population-based TSH reference ranges, were reclassified as euthyroid when genetically determined TSH reference ranges were applied. Individuals in the higher PGS quartiles had a higher probability of being prescribed LT4 treatment compared to individuals from the lower PGS quartiles (3.3% in Q1 vs. 5.2% in Q4, Pfor trend =1.7 × 10-8). Conclusions: Individual genetic profiles have the potential to personalize TSH reference ranges, with large effects on reclassification of diagnosis and LT4 prescriptions. As the currently used PGS can only predict approximately 10% of inter-individual variation in TSH concentrations, it should be further improved when more genetic variants determining TSH concentrations are identified in future studies.
Collapse
Affiliation(s)
- Aleksander Kuś
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine and Endocrinology, Medical University of Warsaw, Warsaw, Poland
| | - Rosalie B T M Sterenborg
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Eirin B Haug
- Department of Public Health and Nursing, HUNT Center for Molecular and Clinical Epidemiology, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - Tessel E Galesloot
- Department for Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands
| | - W Edward Visser
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Johannes W A Smit
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tomasz Bednarczuk
- Department of Internal Medicine and Endocrinology, Medical University of Warsaw, Warsaw, Poland
| | - Robin P Peeters
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Bjørn O Åsvold
- Department of Public Health and Nursing, HUNT Center for Molecular and Clinical Epidemiology, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Endocrinology, Clinic of Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Department of Public Health and Nursing, HUNT Research Centre, NTNU, Norwegian University of Science and Technology, Levanger, Norway
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Bialystok, Bialystok, Poland
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Marco Medici
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
31
|
Mousavi I, Suffredini J, Virani SS, Ballantyne C, Michos ED, Misra A, Saeed A, Jia X. Early Onset Atherosclerotic Cardiovascular Disease. Eur J Prev Cardiol 2024:zwae240. [PMID: 39041374 DOI: 10.1093/eurjpc/zwae240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/16/2024] [Accepted: 07/20/2024] [Indexed: 07/24/2024]
Abstract
Recent trends indicate a concerning increase in early-onset atherosclerotic cardiovascular disease (ASCVD) among younger individuals (age < 55 in men and <65 in women). These findings highlight the pathobiology of ASCVD as a disease process that begins early in life and underscores the need for more tailored screening methods and preventive strategies. Increasing attention has been placed on the growing burden of traditional cardiometabolic risk factors in young individuals while also recognizing unique factors that mediate risk of premature atherosclerosis in this demographic such as substance use, socioeconomic disparities, adverse pregnancy outcomes, and chronic inflammatory states that contribute to the increasing incidence of early ASCVD. Additionally, mounting evidence has pointed out significant disparities in the diagnosis and management of early ASCVD and cardiovascular outcomes based on sex and race. Moving toward a more personalized approach, emerging data and technological developments using diverse tools such as polygenic risk scores and coronary artery calcium scans have shown potential in earlier detection of ASCVD risk. Thus, we review current evidence on causal risk factors that drive the increase in early ASCVD and highlight emerging tools to improve ASCVD risk assessment in young individuals.
Collapse
Affiliation(s)
- Idine Mousavi
- Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - John Suffredini
- Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Salim S Virani
- The Aga Khan University, Karachi, Pakistan; Baylor College of Medicine and Texas Heart Institute, Houston, TX, USA
| | - Christie Ballantyne
- Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Erin D Michos
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Arunima Misra
- Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Section of Cardiology, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
| | - Anum Saeed
- Heart and Vascular Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Xiaoming Jia
- Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
32
|
Woods E, Bennett J, Chandrasekhar S, Newman N, Rizwan A, Siddiqui R, Khan R, Khawaja M, Krittanawong C. Efficacy of Diagnostic Testing of Suspected Coronary Artery Disease: A Contemporary Review. Cardiology 2024:1-22. [PMID: 39013364 DOI: 10.1159/000539916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 06/10/2024] [Indexed: 07/18/2024]
Abstract
BACKGROUND Coronary artery disease (CAD) is a highly prevalent condition which can lead to myocardial ischemia as well as acute coronary syndrome. Early diagnosis of CAD can improve patient outcomes through guiding risk factor modification and treatment modalities. SUMMARY Testing for CAD comes with increased cost and risk; therefore, physicians must determine which patients require testing, and what testing modality will offer the most useful data to diagnose patients with CAD. Patients should have an initial risk stratification for pretest probability of CAD based on symptoms and available clinical data. Patients with a pretest probability less than 5% should receive no further testing, while patients with a high pretest probability should be considered for direct invasive coronary angiography. In patients with a pretest probability between 5 and 15%, coronary artery calcium score and or exercise electrocardiogram can be obtained to further risk stratify patients to low-risk versus intermediate-high-risk. Intermediate-high-risk patients should be tested with coronary computed tomography angiography (preferred) versus positron emission tomography or single photon emission computed tomography based on their individual patient characteristics and institutional availability. KEY MESSAGES This comprehensive review aimed to describe the available CAD testing modalities, detail their risks and benefits, and propose when each should be considered in the evaluation of a patient with suspected CAD.
Collapse
Affiliation(s)
- Edward Woods
- Department of Internal Medicine, Emory University, Atlanta, Georgia, USA
| | - Josiah Bennett
- Department of Internal Medicine, Emory University, Atlanta, Georgia, USA
| | | | - Noah Newman
- Department of Internal Medicine, Emory University, Atlanta, Georgia, USA
| | - Affan Rizwan
- Department of Internal Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Rehma Siddiqui
- Department of Internal Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Rabisa Khan
- Department of Anesthesiology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Muzamil Khawaja
- Division of Cardiology, Emory University, Atlanta, Georgia, USA
| | - Chayakrit Krittanawong
- Cardiology Division, NYU Langone Health and NYU School of Medicine, New York, New York, USA
| |
Collapse
|
33
|
Naderian M, Hamed ME, Vaseem AA, Norland K, Dikilitas O, Teymourzadeh A, Bailey KR, Kullo IJ. Effect of disclosing a polygenic risk score for coronary heart disease on adverse cardiovascular events: 10-year follow-up of the MI-GENES randomized clinical trial. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.19.24310709. [PMID: 39072039 PMCID: PMC11275655 DOI: 10.1101/2024.07.19.24310709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Background The MI-GENES clinical trial (NCT01936675), in which participants at intermediate risk of coronary heart disease (CHD) were randomized to receive a Framingham risk score (FRSg, n=103), or an integrated risk score (IRSg, n=104) that additionally included a polygenic risk score (PRS), demonstrated that after 6 months, participants randomized to IRSg had higher statin initiation and lower low-density lipoprotein cholesterol (LDL-C). Objectives In a post hoc 10-year follow-up analysis of the MI-GENES trial, we investigated whether disclosure of a PRS for CHD was associated with a reduction in adverse cardiovascular events. Methods Participants were followed from randomization beginning in October 2013 until September 2023 to ascertain adverse cardiovascular events, testing for CHD, and changes in risk factors, by blinded review of electronic health records. The primary outcome was the time from randomization to the occurrence of the first major adverse cardiovascular event (MACE), defined as cardiovascular death, non-fatal myocardial infarction, coronary revascularization, and non-fatal stroke. Statistical analyses were conducted using Cox proportional hazards regression and linear mixed-effects models. Results We followed all 203 participants who completed the MI-GENES trial, 100 in FRSg and 103 in IRSg (mean age at the end of follow-up: 68.2±5.2, 48% male). During a median follow-up of 9.5 years, 9 MACEs occurred in FRSg and 2 in IRSg (hazard ratio (HR), 0.20; 95% confidence interval (CI), 0.04 to 0.94; P=0.042). In FRSg, 47 (47%) underwent at least one test for CHD, compared to 30 (29%) in IRSg (HR, 0.51; 95% CI, 0.32 to 0.81; P=0.004). IRSg participants had a longer duration of statin therapy during the first four years post-randomization and a greater reduction in LDL-C for up to 3 years post-randomization. No significant differences between the two groups were observed for hemoglobin A1C, systolic and diastolic blood pressures, weight, and smoking cessation rate during follow-up. Conclusions The disclosure of an IRS that included a PRS to individuals at intermediate risk for CHD was associated with a lower incidence of MACE after a decade of follow-up, likely due to a higher rate of initiation and longer duration of statin therapy, leading to lower LDL-C levels.
Collapse
Affiliation(s)
| | - Marwan E. Hamed
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Ali A. Vaseem
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Kristjan Norland
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Ozan Dikilitas
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Azin Teymourzadeh
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Kent R. Bailey
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Iftikhar J. Kullo
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
- Gonda Vascular Center, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
34
|
Kamal S, Jasraj P, Krutika P, Devratsinh P, Maulik K, Dixit D. To Evaluate Efficiency of Various Coronary Artery Disease Risk Scores With Traditional Risk Factors in Patients Undergoing Coronary Angiography. J Saudi Heart Assoc 2024; 36:128-136. [PMID: 39011030 PMCID: PMC11249062 DOI: 10.37616/2212-5043.1386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 06/03/2024] [Accepted: 06/19/2024] [Indexed: 07/17/2024] Open
Abstract
Objective To analyze and compare various cardiovascular disease risk scores in Western Indian patients undergoing Coronary angiogram (CAG). Methods In this prospective cross-sectional study, 1213 patients who underwent conventional coronary angiography; clinical risk profile and biochemical investigations were evaluated prior to undergoing CAG. Apart from the demographic information, 10-year absolute risk of having a major cardiovascular event (cardiovascular death, myocardial infarction or stroke) was calculated for each patient using various available Traditional Risk Scores (TRS). The population was divided in low, intermediate and high-risk categories for each of these scores. Results Traditional cardiovascular risk factors like hypertension (41.8%) and diabetes mellitus-II (26.9%) were the two most prevalent risk factors in our study population. A higher risk value for all these TRS was more likely to be associated with obstructive coronary artery disease (OCAD) on CAG. Patients with high risk (≥20% for 10-year) QRESEARCH (QRISK3) score category had higher number of patients with obstructive CAD (49.6%) as compared to high risk category of risk score for those with high Global Registry of Acute Coronary Events (GRACE) score (46.6%) or risk Framingham (FRS CHD) score (29.2%) and risk atherosclerotic cardiovascular disease (ASCVD) score (30.1%) (P < 0.0001). A higher TRS was more likely to be associated with obstructive CAD, with the highest predictability being with QRISK3 (QRISK3 score 60.9%, GRACE score 54.9%, FRS-CHD score 34% and ASCVD score 42.1% respectively; P < 0.0001). A substantial study population (27.4%) cannot be identified using any of these TRS and hence a need of indigenous or modified risk scores is proposed. Conclusion QRISK3 score was most efficacious for predicting obstructive CAD in our Indian study population on CAG. A higher risk score also correlated with the number of vessels involved on coronary angiogram. A substantial obstructive CAD patient could not be identified using traditional risk scores hence need for an indigenous or modified score.
Collapse
Affiliation(s)
- Sharma Kamal
- Department of Cardiology, U. N. Mehta Institute of Cardiology and Research Centre (UNMICRC), Civil Hospital Campus, Asarwa, Ahmedabad, 380016, Gujarat, India
| | - Panwar Jasraj
- Interventional Cardiologist, Department of Cardiology, SMVS Hospital, Ahmedabad, Gujarat, India
| | - Patel Krutika
- Department of Research, U. N. Mehta Institute of Cardiology and Research Centre (UNMICRC), Civil Hospital Campus, Asarwa, Ahmedabad, 380016, Gujarat, India
| | - Parmar Devratsinh
- Department of Cardiology, U. N. Mehta Institute of Cardiology and Research Centre (UNMICRC), Civil Hospital Campus, Asarwa, Ahmedabad, 380016, Gujarat, India
| | - Kalyani Maulik
- Department of Cardiology, U. N. Mehta Institute of Cardiology and Research Centre (UNMICRC), Civil Hospital Campus, Asarwa, Ahmedabad, 380016, Gujarat, India
| | - Dhorajiya Dixit
- Department of Cardiology, U. N. Mehta Institute of Cardiology and Research Centre (UNMICRC), Civil Hospital Campus, Asarwa, Ahmedabad, 380016, Gujarat, India
| |
Collapse
|
35
|
Hou K, Xu Z, Ding Y, Mandla R, Shi Z, Boulier K, Harpak A, Pasaniuc B. Calibrated prediction intervals for polygenic scores across diverse contexts. Nat Genet 2024; 56:1386-1396. [PMID: 38886587 PMCID: PMC11465192 DOI: 10.1038/s41588-024-01792-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 05/08/2024] [Indexed: 06/20/2024]
Abstract
Polygenic scores (PGS) have emerged as the tool of choice for genomic prediction in a wide range of fields. We show that PGS performance varies broadly across contexts and biobanks. Contexts such as age, sex and income can impact PGS accuracy with similar magnitudes as genetic ancestry. Here we introduce an approach (CalPred) that models all contexts jointly to produce prediction intervals that vary across contexts to achieve calibration (include the trait with 90% probability), whereas existing methods are miscalibrated. In analyses of 72 traits across large and diverse biobanks (All of Us and UK Biobank), we find that prediction intervals required adjustment by up to 80% for quantitative traits. For disease traits, PGS-based predictions were miscalibrated across socioeconomic contexts such as annual household income levels, further highlighting the need of accounting for context information in PGS-based prediction across diverse populations.
Collapse
Affiliation(s)
- Kangcheng Hou
- Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA.
| | - Ziqi Xu
- Department of Computer Science, University of California Los Angeles, Los Angeles, CA, USA
| | - Yi Ding
- Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA
| | - Ravi Mandla
- Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA
| | - Zhuozheng Shi
- Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA
| | - Kristin Boulier
- Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA
| | - Arbel Harpak
- Department of Population Health, The University of Texas at Austin, Austin, TX, USA
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX, USA
| | - Bogdan Pasaniuc
- Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA.
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Department of Computational Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Institute for Precision Health, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
36
|
Paruchuri K, Bhukar R, Urbut S, Ruan Y, Ganesh S, Postupaka D, Natarajan P. Using Sex-Specific Polygenic Risk to Prognosticate Coronary Artery Disease in Women. J Am Heart Assoc 2024; 13:e034946. [PMID: 38874071 PMCID: PMC11255742 DOI: 10.1161/jaha.123.034946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 05/02/2024] [Indexed: 06/15/2024]
Affiliation(s)
- Kaavya Paruchuri
- Cardiovascular Research CenterMassachusetts General HospitalBostonMA
- Program in Medical and Population Genetics and Cardiovascular Disease InitiativeBroad Institute of Harvard and MITCambridgeMA
- Department of MedicineHarvard Medical SchoolBostonMA
| | - Rohan Bhukar
- Cardiovascular Research CenterMassachusetts General HospitalBostonMA
- Program in Medical and Population Genetics and Cardiovascular Disease InitiativeBroad Institute of Harvard and MITCambridgeMA
| | - Sarah Urbut
- Cardiovascular Research CenterMassachusetts General HospitalBostonMA
- Program in Medical and Population Genetics and Cardiovascular Disease InitiativeBroad Institute of Harvard and MITCambridgeMA
- Department of MedicineHarvard Medical SchoolBostonMA
- Center for Genomic MedicineMassachusetts General HospitalBostonMA
| | - Yunfeng Ruan
- Program in Medical and Population Genetics and Cardiovascular Disease InitiativeBroad Institute of Harvard and MITCambridgeMA
| | - Shriienidhie Ganesh
- Cardiovascular Research CenterMassachusetts General HospitalBostonMA
- Program in Medical and Population Genetics and Cardiovascular Disease InitiativeBroad Institute of Harvard and MITCambridgeMA
| | - Darina Postupaka
- Cardiovascular Research CenterMassachusetts General HospitalBostonMA
- Program in Medical and Population Genetics and Cardiovascular Disease InitiativeBroad Institute of Harvard and MITCambridgeMA
| | - Pradeep Natarajan
- Cardiovascular Research CenterMassachusetts General HospitalBostonMA
- Program in Medical and Population Genetics and Cardiovascular Disease InitiativeBroad Institute of Harvard and MITCambridgeMA
- Department of MedicineHarvard Medical SchoolBostonMA
- Center for Genomic MedicineMassachusetts General HospitalBostonMA
| |
Collapse
|
37
|
Le A, Peng H, Golinsky D, Di Scipio M, Lali R, Paré G. What Causes Premature Coronary Artery Disease? Curr Atheroscler Rep 2024; 26:189-203. [PMID: 38573470 DOI: 10.1007/s11883-024-01200-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2024] [Indexed: 04/05/2024]
Abstract
PURPOSE OF REVIEW This review provides an overview of genetic and non-genetic causes of premature coronary artery disease (pCAD). RECENT FINDINGS pCAD refers to coronary artery disease (CAD) occurring before the age of 65 years in women and 55 years in men. Both genetic and non-genetic risk factors may contribute to the onset of pCAD. Recent advances in the genetic epidemiology of pCAD have revealed the importance of both monogenic and polygenic contributions to pCAD. Familial hypercholesterolemia (FH) is the most common monogenic disorder associated with atherosclerotic pCAD. However, clinical overreliance on monogenic genes can result in overlooked genetic causes of pCAD, especially polygenic contributions. Non-genetic factors, notably smoking and drug use, are also important contributors to pCAD. Cigarette smoking has been observed in 25.5% of pCAD patients relative to 12.2% of non-pCAD patients. Finally, myocardial infarction (MI) associated with spontaneous coronary artery dissection (SCAD) may result in similar clinical presentations as atherosclerotic pCAD. Recognizing the genetic and non-genetic causes underlying pCAD is important for appropriate prevention and treatment. Despite recent progress, pCAD remains incompletely understood, highlighting the need for both awareness and research.
Collapse
Affiliation(s)
- Ann Le
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada
- Department of Medical Sciences, Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
| | - Helen Peng
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada
- Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, L8L 4K1, Canada
| | - Danielle Golinsky
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada
- School of Nursing, Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, L8L 4K1, Canada
| | - Matteo Di Scipio
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada
- Department of Medical Sciences, Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
- Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, ON, L8L 4K1, Canada
| | - Ricky Lali
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada
- Department of Health Research Methods, Evidence, and Impact, McMaster University, 1280 Main Street West, Hamilton, ON, L8L 4K1, Canada
| | - Guillaume Paré
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada.
- Department of Medical Sciences, Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada.
- Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada.
- Thrombosis and Atherosclerosis Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada.
- Department of Pathology and Molecular Medicine, Michael G. DeGroote School of Medicine, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada.
- Department of Health Research Methods, Evidence, and Impact, McMaster University, 1280 Main Street West, Hamilton, ON, L8L 4K1, Canada.
| |
Collapse
|
38
|
Iribarren C, Lu M, Elosua R, Gulati M, Wong ND, Blumenthal RS, Nissen S, Rana JS. Polygenic risk and incident coronary heart disease in a large multiethnic cohort. Am J Prev Cardiol 2024; 18:100661. [PMID: 38601895 PMCID: PMC11004687 DOI: 10.1016/j.ajpc.2024.100661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/08/2024] [Accepted: 03/25/2024] [Indexed: 04/12/2024] Open
Abstract
Objective Many studies support the notion that polygenic risk scores (PRS) improve risk prediction for coronary heart disease (CHD) beyond conventional risk factors. However, PRS are not yet considered risk-enhancing factor in guidelines. Our objective was to determine the predictive performance of a commercially available PRS (CARDIO inCode-Score®) compared with the Pooled Cohorts Equations (PCE) in a contemporary, multi-ethnic cohort. Methods Participants (n = 63,070; 67 % female; 18 % non-European) without prior CHD were followed from 2007 through 12/31/2022. The association between the PRS and incident CHD was assessed using Cox regression adjusting for genetic ancestry and risk factors. Event rates were estimated by categories of PCE and by low/intermediate/high genetic risk within PCE categories; risk discrimination and net reclassification improvement (NRI) were also assessed. Results There were 3,289 incident CHD events during 14 years of follow-up. Adjusted hazard ratio (aHR) for incident CHD per 1 SD increase in PRS was 1.18 (95 % CI:1.14-1.22), and the aHR for the upper vs lower quintile of the PRS was 1.66 (95 % CI:1.49-1.86). The association was consistent in both sexes, in European participants compared with all minority groups combined and was strongest in the first 5 years of follow-up. The increase in the C-statistic was 0.004 (0.747 vs. 0.751; p < 0.0001); the NRI was 2.4 (0.9-3.8) for the entire cohort and 9.7 (7.5-12.0) for intermediate PCE risk individuals. After incorporating high genetic risk, a further 10 percent of participants at borderline/intermediate PCE risk would be candidates for statin therapy. Conclusion Inclusion of polygenic risk improved identification of primary prevention individuals who may benefit from more intensive risk factor modification.
Collapse
Affiliation(s)
- Carlos Iribarren
- Kaiser Permanente Northern California Division of Research, Oakland, CA, USA
| | - Meng Lu
- Kaiser Permanente Northern California Division of Research, Oakland, CA, USA
| | - Roberto Elosua
- Cardiovascular Epidemiology and Genetics, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Spain and CIBER Cardiovascular Diseases (CIBERCV), Barcelona, Spain
- Faculty of Medicine, University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain
| | - Martha Gulati
- Barbra Streisand Women's Heart Center, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Nathan D. Wong
- Heart Disease Prevention Program, Division of Cardiology, Department of Medicine, University of California Irvine, Irvine, CA, USA
| | - Roger S. Blumenthal
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Steven Nissen
- Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Jamal S. Rana
- Kaiser Permanente Northern California Division of Research, Oakland, CA, USA
- Department of Cardiology, The Permanente Medical Group, Kaiser Permanente Oakland Medical Center, Oakland, CA, USA
| |
Collapse
|
39
|
Henry J, Lin Y, Bouatia-Naji N. Enhancing the Prediction Power of Polygenic Risk Scores in Genetically Diverse Coronary Heart Disease. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2024; 17:e004610. [PMID: 38586952 DOI: 10.1161/circgen.124.004610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Affiliation(s)
- Joséphine Henry
- Université Paris Cité, Paris-Cardiovascular Research Center, Institut National de la Sante et de la Recherche Medicale, France
| | - Yilong Lin
- Université Paris Cité, Paris-Cardiovascular Research Center, Institut National de la Sante et de la Recherche Medicale, France
| | - Nabila Bouatia-Naji
- Université Paris Cité, Paris-Cardiovascular Research Center, Institut National de la Sante et de la Recherche Medicale, France
| |
Collapse
|
40
|
Ranjan R, Hasan MK, Adhikary AB. Bangladeshi Atherosclerosis Biobank and Hub: The BANGABANDHU Study. Int J Gen Med 2024; 17:2507-2512. [PMID: 38826511 PMCID: PMC11144007 DOI: 10.2147/ijgm.s466706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 05/24/2024] [Indexed: 06/04/2024] Open
Abstract
Background Genetic factors contribute significantly to the risk of ischaemic heart disease (IHD), which is the leading cause of mortality in Bangladesh. The BANGABANDHU (Bangladeshi Atherosclerosis Biobank AND Hub) study will allow a hypothesis-free genome-wide association study (GWAS) to identify genetic risk factors associated with ischaemic heart disease patients undergoing coronary artery bypass graft (CABG) surgery in Bangladesh. Methods This is a multi-centre population-based case-control study aimed to evaluate 1500 (Fifteen Hundred) adult (≥18 years of age) people divided into 2 study groups: Case/Proband (750 IHD patients undergoing CABG surgery) and Control (750 healthy people). Spouses or family members are preferred as healthy control subjects due to their shared geographic location and similar environmental exposure. Results This will be the first largest DNA repository of CABG patients in Bangladesh, and identifying novel gene loci among CABG patients might help to discover novel therapeutic targets for Bangladeshi IHD patients. Further, identifying and comparing novel gene loci among CABG patients with other ancestry might help devise national guidelines for treating coronary artery disease. Conclusion Promising current study results will encourage Bangladeshi researchers and pharmaceutical companies to conduct further studies into the genetic basis of Bangladeshi complex coronary artery disease, which might identify novel genes for therapeutic targets for Bangladeshi patients and strengthen the healthcare standards in Bangladesh.
Collapse
Affiliation(s)
- Redoy Ranjan
- Department of Cardiac Surgery, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - Md Kamrul Hasan
- Department of Cardiac Surgery, National Institute of Cardiovascular Diseases, Dhaka, Bangladesh
| | - Asit Baran Adhikary
- Department of Cardiac Surgery, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
- Department of Cardiac Surgery, Impulse Hospital & Research Centre, Dhaka, Bangladesh
| |
Collapse
|
41
|
Nielsen RV, Fuster V, Bundgaard H, Fuster JJ, Johri AM, Kofoed KF, Douglas PS, Diederichsen A, Shapiro MD, Nicholls SJ, Nordestgaard BG, Lindholt JS, MacRae C, Yuan C, Newby DE, Urbina EM, Bergström G, Ridderstråle M, Budoff MJ, Bøttcher M, Raitakari OT, Hansen TH, Näslund U, Sillesen H, Eldrup N, Ibanez B. Personalized Intervention Based on Early Detection of Atherosclerosis: JACC State-of-the-Art Review. J Am Coll Cardiol 2024; 83:2112-2127. [PMID: 38777513 DOI: 10.1016/j.jacc.2024.02.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/12/2024] [Accepted: 02/22/2024] [Indexed: 05/25/2024]
Abstract
Cardiovascular disease (CVD) remains the leading cause of morbidity and mortality worldwide and challenges the capacity of health care systems globally. Atherosclerosis is the underlying pathophysiological entity in two-thirds of patients with CVD. When considering that atherosclerosis develops over decades, there is potentially great opportunity for prevention of associated events such as myocardial infarction and stroke. Subclinical atherosclerosis has been identified in its early stages in young individuals; however, there is no consensus on how to prevent progression to symptomatic disease. Given the growing burden of CVD, a paradigm shift is required-moving from late management of atherosclerotic CVD to earlier detection during the subclinical phase with the goal of potential cure or prevention of events. Studies must focus on how precision medicine using imaging and circulating biomarkers may identify atherosclerosis earlier and determine whether such a paradigm shift would lead to overall cost savings for global health.
Collapse
Affiliation(s)
- Rikke V Nielsen
- Department of Medical Science, Novo Nordisk Foundation, Hellerup, Denmark; Department of Cardiothoracic Anesthesiology, Rigshospitalet University Hospital Copenhagen, Copenhagen, Denmark.
| | - Valentin Fuster
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Mount Sinai Fuster Heart Hospital, New York, New York, USA
| | - Henning Bundgaard
- Department of Cardiology, Rigshospitalet University Hospital Copenhagen, Copenhagen, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jose J Fuster
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; CIBER en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Amer M Johri
- Department of Medicine Queen's University, Kingston, Ontario, Canada
| | - Klaus F Kofoed
- Department of Cardiology, Rigshospitalet University Hospital Copenhagen, Copenhagen, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Radiology, Rigshospitalet University Hospital Copenhagen, Copenhagen, Denmark
| | - Pamela S Douglas
- Duke University School of Medicine, Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Axel Diederichsen
- Department of Cardiology, Odense University Hospital, Odense, Denmark
| | - Michael D Shapiro
- Center for Prevention of Cardiovascular Disease, Section on Cardiovascular Disease, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Stephen J Nicholls
- Victorian Heart Institute, Monash University, Melbourne, Victoria, Australia
| | - Børge G Nordestgaard
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Biochemistry and The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark. https://twitter.com/BNordestgaard
| | - Jes S Lindholt
- Department of Cardiothoracic and Vascular Surgery, Elite Research Centre of Individualised Treatment of Arterial Disease (CIMA), Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Calum MacRae
- Harvard Medical School, Department of Medicine, Boston, Massachusetts, USA
| | - Chun Yuan
- Department of Radiology and Imaging Sciences, Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - David E Newby
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, Scotland
| | - Elaine M Urbina
- Preventive Cardiology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati, Cincinnati, Ohio, USA
| | - Göran Bergström
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg and Department of Clinical Physiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | - Matthew J Budoff
- Department of Medicine, Lundquist Institute at Harbor-UCLA, Torrance, California, USA
| | - Morten Bøttcher
- University Clinic for Cardiovascular Research, Department of Cardiology, Aarhus University/Gødstrup Hospital, Aarhus, Denmark
| | - Olli T Raitakari
- Centre for Population Health Research, Research Centre of Applied and Preventive Cardiovascular Medicine, InFLAMES Research Flagship, University of Turku, Turku, Finland; Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| | - Thomas H Hansen
- Department of Cardiology, Rigshospitalet University Hospital Copenhagen, Copenhagen, Denmark
| | - Ulf Näslund
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Henrik Sillesen
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nikolaj Eldrup
- Department of Vascular Surgery, Rigshospitalet University Hospital Copenhagen, Copenhagen, Denmark
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; CIBER en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain; Cardiology Department, IIS-Fundación Jiménez Díaz University Hospital, Madrid, Spain.
| |
Collapse
|
42
|
Natarajan P, Bellomo TR. Clonal Hematopoiesis Among Patients With Asymptomatic Carotid Stenosis Compounds Risk of Cardiovascular Death. J Am Coll Cardiol 2024; 83:1728-1730. [PMID: 38692826 DOI: 10.1016/j.jacc.2024.03.389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 03/19/2024] [Accepted: 03/19/2024] [Indexed: 05/03/2024]
Affiliation(s)
- Pradeep Natarajan
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA; Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA.
| | - Tiffany R Bellomo
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA; Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA; Division of Vascular and Endovascular Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA. https://twitter.com/BellomoTiffany
| |
Collapse
|
43
|
Puri R, Bansal M, Mehta V, Duell PB, Wong ND, Iyengar SS, Kalra D, Nair DR, Nanda NC, Narula J, Deedwania P, Yusuf J, Dalal JJ, Shetty S, Vijan VM, Agarwala R, Kumar S, Vijay K, Khan A, Wander GS, Manoria PC, Wangnoo SK, Mohan V, Joshi SR, Singh B, Kerkar P, Rajput R, Prabhakar D, Zargar AH, Saboo B, Kasliwal RR, Ray S, Bansal S, Rabbani MU, Chhabra ST, Chandra S, Bardoloi N, Kavalipati N, Sathyamurthy I, Mahajan K, Pradhan A, Khanna NN, Khadgawat R, Gupta P, Chag MC, Gupta A, Murugnathan A, Narasingan SN, Upadhyaya S, Mittal V, Melinkeri RP, Yadav M, Mubarak MR, Pareek KK, Dabla PK, Nanda R, Mohan JC. Lipid Association of India 2023 update on cardiovascular risk assessment and lipid management in Indian patients: Consensus statement IV. J Clin Lipidol 2024; 18:e351-e373. [PMID: 38485619 DOI: 10.1016/j.jacl.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/16/2024] [Accepted: 01/25/2024] [Indexed: 06/28/2024]
Abstract
OBJECTIVE In 2016, the Lipid Association of India (LAI) developed a cardiovascular risk assessment algorithm and defined low-density lipoprotein cholesterol (LDL-C) goals for prevention of atherosclerotic cardiovascular disease (ASCVD) in Indians. The recent refinements in the role of various risk factors and subclinical atherosclerosis in prediction of ASCVD risk necessitated updating the risk algorithm and treatment goals. METHODS The LAI core committee held twenty-one meetings and webinars from June 2022 to July 2023 with experts across India and critically reviewed the latest evidence regarding the strategies for ASCVD risk prediction and the benefits and modalities for intensive lipid lowering. Based on the expert consensus and extensive review of published data, consensus statement IV was commissioned. RESULTS The young age of onset and a more aggressive nature of ASCVD in Indians necessitates emphasis on lifetime ASCVD risk instead of the conventional 10-year risk. It also demands early institution of aggressive preventive measures to protect the young population prior to development of ASCVD events. Wide availability and low cost of statins in India enable implementation of effective LDL-C-lowering therapy in individuals at high risk of ASCVD. Subjects with any evidence of subclinical atherosclerosis are likely to benefit the most from early aggressive interventions. CONCLUSIONS This document presents the updated risk stratification and treatment algorithm and describes the rationale for each modification. The intent of these updated recommendations is to modernize management of dyslipidemia in Indian patients with the goal of reducing the epidemic of ASCVD among Indians in Asia and worldwide.
Collapse
Affiliation(s)
- Raman Puri
- Chair, FNLA, Sr. Consultant Cardiologist, Cardiac Care Centre, New Delhi, India (Dr Puri).
| | - Manish Bansal
- Co-Chair, Senior Director, Department of Cardiology, Medanta- The Medicity, Gurugram, Haryana, India (Dr Bansal)
| | - Vimal Mehta
- Co-Chair, Director-Professor, Department of Cardiology, G. B. Pant Institute of Postgraduate Medical Education and Research, New Delhi, India (Dr Mehta)
| | - P Barton Duell
- Co-Chair, FNLA, Professor of Medicine, Knight Cardiovascular Institute and Division of Endocrinology Diabetes and Clinical Nutrition, Oregon Health & Science University, Portland, OR, USA (Dr Duell)
| | - Nathan D Wong
- FNLA, Professor & Director Heart Disease Prevention program division of Cardiology, University of California, Irvine School of Medicine, USA (Dr Wong)
| | - S S Iyengar
- Sr. Consultant and Head, Department of Cardiology, Manipal Hospital, Bangalore, Karnataka, India (Dr Iyengar)
| | - Dinesh Kalra
- FNLA, Professor of Medicine, University of Louisville School of Medicine, USA (Dr Kalra)
| | - Devaki R Nair
- Sr. Consultant Department of Lipidology and Chemical pathologist, Royal Free Hospital, London, UK (Dr Nair)
| | - Navin C Nanda
- Professor of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, KY, USA (Dr Nanda)
| | - Jagat Narula
- Executive Vice President and Chief Academic Officer, UT Health, Houston, TX USA (Dr Narula)
| | - P Deedwania
- Professor of Medicine, University of California San Francisco, San Francisco, CA, USA (Dr Deedwania)
| | - Jamal Yusuf
- Director-Professor and Head, Department of Cardiology, G. B. Pant Institute of Postgraduate Medical Education and Research, New Delhi, India (Dr Yusuf)
| | - Jamshed J Dalal
- Sr. Consultant Cardiologist, Kokilaben Dhirubhai Ambani Hospital, Director-Centre for Cardiac Sciences, Mumbai, Maharashtra, India (Dr Dalal)
| | - Sadanand Shetty
- Head, Department of Cardiology, K. J. Somaiya Super Specialty Institute, Sion (East), Mumbai, Maharashtra, India (Dr Shetty)
| | - Vinod M Vijan
- Director, Vijan Hospital & Research Centre, Nashik, Uniqare Hospital, PCMC, Pune, India (Dr Vijan)
| | - Rajeev Agarwala
- Sr. Consultant Cardiologist, Jaswant Rai Specialty Hospital, Meerut, Uttar Pradesh, India (Dr Agarwala)
| | - Soumitra Kumar
- Professor and Head, Department of Cardiology, Vivekananda Institute of Medical Sciences, Kolkata, India (Dr Kumar)
| | - Kris Vijay
- FNLA, Professor of Medicine, Arizona Heart Foundation, University of Arizona, Phoenix, USA (Dr Vijay)
| | - Aziz Khan
- Sr. Consultant cardiologist, Crescent Hospital and Heart Centre, Nagpur, Maharashtra, India (Dr Khan)
| | - Gurpreet Singh Wander
- Professor of Cardiology, Dayanand Medical College and Hospital, Ludhiana, Punjab, India (Dr Wander)
| | - P C Manoria
- Director, Manoria Heart and critical Care Hospital, Bhopal, Madhya Pradesh, India (Dr Manoria)
| | - S K Wangnoo
- Sr. Consultant Endocrinology & Diabetologist, Indraprastha Apollo Hospitals, New Delhi, India (Dr Wangnoo)
| | - Viswanathan Mohan
- Director Madras Diabetic Research foundation and Chairman & chief Diabetology, Dr Mohan Diabetes Specialties Centre, Chennai, India (Dr Mohan)
| | - Shashank R Joshi
- Sr. Consultant Endocrinologist, Lilavati Hospital, Mumbai, Maharashtra, India (Dr Joshi)
| | - Balbir Singh
- Chairman - Cardiac Sciences, Max Hospital Saket, New Delhi, India (Dr Singh)
| | - Prafulla Kerkar
- Sr. Consultant Cardiologist, Asian Heart Institute and Research Centre, Mumbai, India (Dr Kerkar)
| | - Rajesh Rajput
- Professor & Head, Department of Endocrinology, Post Graduate Institute of Medical Sciences, Rohtak, Haryana, India (Dr Rajput)
| | - D Prabhakar
- Sr. Consultant, Department of Cardiology, Apollo Hospitals, Chennai, Tamil Nadu, India (Dr Prabhakar)
| | - Abdul Hamid Zargar
- Medical Director, Centre for Diabetes and Endocrine Care, National Highway, Gulshan Nagar, Srinagar, J&K, India (Dr Zargar)
| | - Banshi Saboo
- Chairman-Diacare- Diabetes Care, and Hormone Clinic, Ahmedabad, India (Dr Saboo)
| | - Ravi R Kasliwal
- Chairman, Division of Clinical & Preventive Cardiology, Medanta- The Medicity, Gurugram, Haryana, India (Dr Kasliwal)
| | - Saumitra Ray
- Director of Intervention Cardiology, AMRI (S), Kolkata, India (Dr Ray)
| | - Sandeep Bansal
- Professor and Head, Dept. of Cardiology, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India (Dr Bansal)
| | - M U Rabbani
- Professor Dept. of Cardiology, J. N. Medical College, AMU, Aligarh, India (Dr Rabbani)
| | - Shibba Takkar Chhabra
- Professor Dept. of Cardiology, Dayanand Medical College and Hospital, Ludhiana, India (Dr Chhabra)
| | - Sarat Chandra
- Chief Cardiologist, TX Group of Hospitals, Banjara Hills, Hyderabad, India (Dr Chandra)
| | - Neil Bardoloi
- Managing Director and HOD, Cardiology, Excel Care Hospital, Guwahati, Assam, India (Dr Bardoloi)
| | - Narasaraju Kavalipati
- Director of Cardiology and Sr Interventional Cardiologist, Apollo Hospitals, Hyderabad, India (Dr Kavalipati)
| | - Immaneni Sathyamurthy
- Sr. Consultant Cardiologist, Apollo Hospital, Chennai, Tamil Nadu, India (Dr Sathyamurthy)
| | - Kunal Mahajan
- Director Dept. of Cardiology, Himachal Heart Institute, Mandi, Himachal Pradesh, India (Dr Mahajan)
| | - Akshya Pradhan
- Sr. Consultant, Department of Cardiology King George's Medical University, Lucknow, Uttar Pradesh, India (Dr Pradhan)
| | - N N Khanna
- Sr. Consultant, Department of Cardiology, Indraprastha Apollo Hospitals, New Delhi, India (Dr Khanna)
| | - Rajesh Khadgawat
- Professor, Department of Endocrinology and Metabolism, All India Institute of Medical Sciences (AIIMS), New Delhi, India (Dr Khadgawat)
| | - Preeti Gupta
- Associate Professor Dept. of Cardiology, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India (Dr Gupta)
| | - Milan C Chag
- Sr. Consultant Cardiologist, Marengo CIMS Hospital, Ahmadabad, Gujarat, India (Dr Chag)
| | - Ashu Gupta
- Sr Consultant Cardiologist, Holy Heart Advanced Cardiac Care and Research Centre, Rohtak, Haryana, India (Dr Gupta)
| | - A Murugnathan
- Sr. Consultant Internal Medicine, AG Hospital, Tirupur, Tamil Nadu, India (Dr Murugnathan)
| | - S N Narasingan
- Former Adjunct Professor of Medicine, The Tamil Nadu Dr MGR Medical University & Managing Director, SNN Specialties Clinic, Chennai, India (Dr Narasingan)
| | - Sundeep Upadhyaya
- Sr. Consultant, Department of Rheumatology, Indraprastha Apollo Hospitals, New Delhi, India (Dr Upadhyaya)
| | - Vinod Mittal
- Sr. Consultant Diabetologist and Head, Centre for Diabetes & Metabolic disease Delhi Heart & Lung Institute, Delhi, India (Dr Mittal)
| | - Rashida Patanwala Melinkeri
- Sr. Consultant, Department of Internal Medicine, KEM Hospital and Sahyadri Hospitals, Pune, Maharashtra, India (Dr Melinkeri)
| | - Madhur Yadav
- Director- Professor of Medicine, Lady Harding Medical College, New Delhi, India (Dr Yadav)
| | - M Raseed Mubarak
- Sr. Consultant Cardiologist, Lanka Hospital, Colombo, Sri Lanka (Dr Mubarak)
| | - K K Pareek
- Head, Department of Medicine, S. N. Pareek Hospital, Dadabari, Kota, Rajasthan, India (Dr Pareek)
| | - Pradeep Kumar Dabla
- Professor of Biochemistry, G. B. Pant Institute of Postgraduate Medical Education and Research, New Delhi, India (Dr Dabla)
| | - Rashmi Nanda
- Managing Director, Ashakiran Family Wellness Clinic, Indrapuram, U.P, India (Dr Nanda)
| | - J C Mohan
- Sr. Consultant Cardiologist, Institute of Heart and Vascular Diseases, Jaipur Golden Hospital, New Delhi, India (Dr Mohan)
| |
Collapse
|
44
|
Foreman AL, Warth B, Hessel EVS, Price EJ, Schymanski EL, Cantelli G, Parkinson H, Hecht H, Klánová J, Vlaanderen J, Hilscherova K, Vrijheid M, Vineis P, Araujo R, Barouki R, Vermeulen R, Lanone S, Brunak S, Sebert S, Karjalainen T. Adopting Mechanistic Molecular Biology Approaches in Exposome Research for Causal Understanding. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:7256-7269. [PMID: 38641325 PMCID: PMC11064223 DOI: 10.1021/acs.est.3c07961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/21/2024]
Abstract
Through investigating the combined impact of the environmental exposures experienced by an individual throughout their lifetime, exposome research provides opportunities to understand and mitigate negative health outcomes. While current exposome research is driven by epidemiological studies that identify associations between exposures and effects, new frameworks integrating more substantial population-level metadata, including electronic health and administrative records, will shed further light on characterizing environmental exposure risks. Molecular biology offers methods and concepts to study the biological and health impacts of exposomes in experimental and computational systems. Of particular importance is the growing use of omics readouts in epidemiological and clinical studies. This paper calls for the adoption of mechanistic molecular biology approaches in exposome research as an essential step in understanding the genotype and exposure interactions underlying human phenotypes. A series of recommendations are presented to make the necessary and appropriate steps to move from exposure association to causation, with a huge potential to inform precision medicine and population health. This includes establishing hypothesis-driven laboratory testing within the exposome field, supported by appropriate methods to read across from model systems research to human.
Collapse
Affiliation(s)
- Amy L. Foreman
- European
Molecular Biology Laboratory & European Bioinformatics Institute
(EMBL-EBI), Wellcome Trust Genome Campus, Hinxton CB10 1SD, U.K.
| | - Benedikt Warth
- Department
of Food Chemistry and Toxicology, University
of Vienna, 1090 Vienna, Austria
| | - Ellen V. S. Hessel
- National
Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA Bilthoven, The Netherlands
| | - Elliott J. Price
- RECETOX,
Faculty of Science, Masaryk University, Kotlarska 2, Brno 60200, Czech Republic
| | - Emma L. Schymanski
- Luxembourg
Centre for Systems Biomedicine, University
of Luxembourg, 6 avenue
du Swing, L-4367 Belvaux, Luxembourg
| | - Gaia Cantelli
- European
Molecular Biology Laboratory & European Bioinformatics Institute
(EMBL-EBI), Wellcome Trust Genome Campus, Hinxton CB10 1SD, U.K.
| | - Helen Parkinson
- European
Molecular Biology Laboratory & European Bioinformatics Institute
(EMBL-EBI), Wellcome Trust Genome Campus, Hinxton CB10 1SD, U.K.
| | - Helge Hecht
- RECETOX,
Faculty of Science, Masaryk University, Kotlarska 2, Brno 60200, Czech Republic
| | - Jana Klánová
- RECETOX,
Faculty of Science, Masaryk University, Kotlarska 2, Brno 60200, Czech Republic
| | - Jelle Vlaanderen
- Institute
for Risk Assessment Sciences, Division of Environmental Epidemiology, Utrecht University, Heidelberglaan 8 3584 CS Utrecht, The Netherlands
| | - Klara Hilscherova
- RECETOX,
Faculty of Science, Masaryk University, Kotlarska 2, Brno 60200, Czech Republic
| | - Martine Vrijheid
- Institute
for Global Health (ISGlobal), Barcelona
Biomedical Research Park (PRBB), Doctor Aiguader, 88, 08003 Barcelona, Spain
- Universitat
Pompeu Fabra, Carrer
de la Mercè, 12, Ciutat Vella, 08002 Barcelona, Spain
- Centro de Investigación Biomédica en Red
Epidemiología
y Salud Pública (CIBERESP), Av. Monforte de Lemos, 3-5. Pebellón 11, Planta 0, 28029 Madrid, Spain
| | - Paolo Vineis
- Department
of Epidemiology and Biostatistics, School of Public Health, Imperial College, London SW7 2AZ, U.K.
| | - Rita Araujo
- European Commission, DG Research and Innovation, Sq. Frère-Orban 8, 1000 Bruxelles, Belgium
| | | | - Roel Vermeulen
- Institute
for Risk Assessment Sciences, Division of Environmental Epidemiology, Utrecht University, Heidelberglaan 8 3584 CS Utrecht, The Netherlands
| | - Sophie Lanone
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Creteil, France
| | - Søren Brunak
- Novo
Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Blegdamsvej 3B, 2200 København, Denmark
| | - Sylvain Sebert
- Research
Unit of Population Health, University of
Oulu, P.O. Box 8000, FI-90014 Oulu, Finland
| | - Tuomo Karjalainen
- European Commission, DG Research and Innovation, Sq. Frère-Orban 8, 1000 Bruxelles, Belgium
| |
Collapse
|
45
|
Holla B, Mahadevan J, Ganesh S, Sud R, Janardhanan M, Balachander S, Strom N, Mattheisen M, Sullivan PF, Huang H, Zandi P, Benegal V, Reddy YJ, Jain S, Purushottam M, Viswanath B. A cross ancestry genetic study of psychiatric disorders from India. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.25.24306377. [PMID: 38712191 PMCID: PMC11071591 DOI: 10.1101/2024.04.25.24306377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Genome-wide association studies across diverse populations may help validate and confirm genetic contributions to risk of disease. We estimated the extent of population stratification as well as the predictive accuracy of polygenic scores (PGS) derived from European samples to a data set from India. We analysed 2685 samples from two data sets, a population neurodevelopmental study (cVEDA) and a hospital-based sample of bipolar affective disorder (BD) and obsessive-compulsive disorder (OCD). Genotyping was conducted using Illumina's Global Screening Array. Population structure was examined with principal component analysis (PCA), uniform manifold approximation and projection (UMAP), support vector machine (SVM) ancestry predictions, and admixture analysis. PGS were calculated from the largest available European discovery GWAS summary statistics for BD, OCD, and externalizing traits using two Bayesian methods that incorporate local linkage disequilibrium structures (PGS-CS-auto) and functional genomic annotations (SBayesRC). Our analyses reveal global and continental PCA overlap with other South Asian populations. Admixture analysis revealed a north-south genetic axis within India (FST 1.6%). The UMAP partially reconstructed the contours of the Indian subcontinent. The Bayesian PGS analyses indicates moderate-to-high predictive power for BD. This was despite the cross-ancestry bias of the discovery GWAS dataset, with the currently available data. However, accuracy for OCD and externalizing traits was much lower. The predictive accuracy was perhaps influenced by the sample size of the discovery GWAS and phenotypic heterogeneity across the syndromes and traits studied. Our study results highlight the accuracy and generalizability of newer PGS models across ancestries. Further research, across diverse populations, would help understand causal mechanisms that contribute to psychiatric syndromes and traits.
Collapse
Affiliation(s)
- Bharath Holla
- Department of Integrative Medicine, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Jayant Mahadevan
- Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Suhas Ganesh
- Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Reeteka Sud
- Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Meghana Janardhanan
- Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Srinivas Balachander
- Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Nora Strom
- Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Munich, Germany
| | - Manuel Mattheisen
- Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Munich, Germany
- Dalhousie University, Department of Community Health and Epidemiology & Faculty of Computer Science, Halifax, Nova Scotia, Canada
- University Hospital of Psychiatry and Psychotherapy, University of Bern
| | - Patrick F Sullivan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Genetics and Psychiatry, University of North Carolina, Chapel Hill, NC, USA
| | - Hailiang Huang
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA02114, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA02142, USA
- Department of Medicine, Harvard Medical School, Boston, MA02114, USA
| | - Peter Zandi
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Vivek Benegal
- Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Yc Janardhan Reddy
- Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Sanjeev Jain
- Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Meera Purushottam
- Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Biju Viswanath
- Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bangalore, India
| |
Collapse
|
46
|
Zou RS, Ruan Y, Truong B, Bhattacharya R, Lu MT, Karády J, Bernardo R, Finneran P, Hornsby W, Fitch KV, Ribaudo HJ, Zanni MV, Douglas PS, Grinspoon SK, Patel AP, Natarajan P. Polygenic Scores and Preclinical Cardiovascular Disease in Individuals With HIV: Insights From the REPRIEVE Trial. J Am Heart Assoc 2024; 13:e033413. [PMID: 38533953 PMCID: PMC11179771 DOI: 10.1161/jaha.123.033413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 01/23/2024] [Indexed: 03/28/2024]
Abstract
BACKGROUND Coronary artery disease (CAD) is a leading cause of death among the 38.4 million people with HIV globally. The extent to which cardiovascular polygenic risk scores (PRSs) derived in non-HIV populations generalize to people with HIV is not well understood. METHODS AND RESULTS PRSs for CAD (GPSMult) and lipid traits were calculated in a global cohort of people with HIV treated with antiretroviral therapy with low-to-moderate atherosclerotic cardiovascular disease risk enrolled in REPRIEVE (Randomized Trial to Prevent Vascular Events in HIV). The PRSs were associated with baseline lipid traits in 4495 genotyped participants, and with subclinical CAD in a subset of 662 who underwent coronary computed tomography angiography. Among participants who underwent coronary computed tomography angiography (mean age, 50.9 [SD, 5.8] years; 16.1% women; 41.8% African, 57.3% European, 1.1% Asian), GPSMult was associated with plaque presence with odds ratio (OR) per SD in GPSMult of 1.42 (95% CI, 1.20-1.68; P=3.8×10-5), stenosis >50% (OR, 2.39 [95% CI, 1.48-3.85]; P=3.4×10-4), and noncalcified/vulnerable plaque (OR, 1.45 [95% CI, 1.23-1.72]; P=9.6×10-6). Effects were consistent in subgroups of age, sex, 10-year atherosclerotic cardiovascular disease risk, ancestry, and CD4 count. Adding GPSMult to established risk factors increased the C-statistic for predicting plaque presence from 0.718 to 0.734 (P=0.02). Furthermore, a PRS for low-density lipoprotein cholesterol was associated with plaque presence with OR of 1.21 (95% CI, 1.01-1.44; P=0.04), and partially calcified plaque with OR of 1.21 (95% CI, 1.01-1.45; P=0.04) per SD. CONCLUSIONS Among people with HIV treated with antiretroviral therapy without documented atherosclerotic cardiovascular disease and at low-to-moderate calculated risk in REPRIEVE, an externally developed CAD PRS was predictive of subclinical atherosclerosis. PRS for low-density lipoprotein cholesterol was also associated with subclinical atherosclerosis, supporting a role for low-density lipoprotein cholesterol in HIV-associated CAD. REGISTRATION URL: https://www.reprievetrial.org; Unique identifier: NCT02344290.
Collapse
Affiliation(s)
- Roger S Zou
- Department of Medicine Massachusetts General Hospital Boston MA USA
- Cardiovascular Disease Initiative Broad Institute of MIT and Harvard Cambridge MA USA
- Harvard Medical School Boston MA USA
| | - Yunfeng Ruan
- Cardiovascular Disease Initiative Broad Institute of MIT and Harvard Cambridge MA USA
| | - Buu Truong
- Cardiovascular Disease Initiative Broad Institute of MIT and Harvard Cambridge MA USA
| | - Romit Bhattacharya
- Cardiovascular Disease Initiative Broad Institute of MIT and Harvard Cambridge MA USA
- Harvard Medical School Boston MA USA
- Division of Cardiology, Department of Medicine, Center for Genomic Medicine Massachusetts General Hospital Boston MA USA
| | - Michael T Lu
- Harvard Medical School Boston MA USA
- Cardiovascular Imaging Research Center Massachusetts General Hospital and Harvard Medical School Boston MA USA
| | - Júlia Karády
- Harvard Medical School Boston MA USA
- Cardiovascular Imaging Research Center Massachusetts General Hospital and Harvard Medical School Boston MA USA
| | - Rachel Bernardo
- Division of Cardiology, Department of Medicine, Center for Genomic Medicine Massachusetts General Hospital Boston MA USA
| | - Phoebe Finneran
- Division of Cardiology, Department of Medicine, Center for Genomic Medicine Massachusetts General Hospital Boston MA USA
| | - Whitney Hornsby
- Division of Cardiology, Department of Medicine, Center for Genomic Medicine Massachusetts General Hospital Boston MA USA
| | - Kathleen V Fitch
- Harvard Medical School Boston MA USA
- Metabolism Unit Massachusetts General Hospital Boston MS USA
| | - Heather J Ribaudo
- Department of Biostatistics, Center for Biostatistics in AIDS Research Harvard TH Chan School of Public Health Boston MA USA
| | - Markella V Zanni
- Harvard Medical School Boston MA USA
- Metabolism Unit Massachusetts General Hospital Boston MS USA
| | - Pamela S Douglas
- Duke Clinical Research Institute, Duke University School of Medicine Durham NC USA
| | - Steven K Grinspoon
- Harvard Medical School Boston MA USA
- Metabolism Unit Massachusetts General Hospital Boston MS USA
| | - Aniruddh P Patel
- Cardiovascular Disease Initiative Broad Institute of MIT and Harvard Cambridge MA USA
- Harvard Medical School Boston MA USA
- Division of Cardiology, Department of Medicine, Center for Genomic Medicine Massachusetts General Hospital Boston MA USA
| | - Pradeep Natarajan
- Cardiovascular Disease Initiative Broad Institute of MIT and Harvard Cambridge MA USA
- Harvard Medical School Boston MA USA
- Division of Cardiology, Department of Medicine, Center for Genomic Medicine Massachusetts General Hospital Boston MA USA
| |
Collapse
|
47
|
Singh RK, Zhao Y, Elze T, Fingert J, Gordon M, Kass MA, Luo Y, Pasquale LR, Scheetz T, Segrè AV, Wiggs JL, Zebardast N. Polygenic Risk Scores for Glaucoma Onset in the Ocular Hypertension Treatment Study. JAMA Ophthalmol 2024; 142:356-363. [PMID: 38483402 PMCID: PMC10941023 DOI: 10.1001/jamaophthalmol.2024.0151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 01/14/2024] [Indexed: 03/17/2024]
Abstract
Importance Primary open-angle glaucoma (POAG) is a highly heritable disease, with 127 identified risk loci to date. Polygenic risk score (PRS) may provide a clinically useful measure of aggregate genetic burden and improve patient risk stratification. Objective To assess whether a PRS improves prediction of POAG onset in patients with ocular hypertension. Design, Setting, and Participants This was a post hoc analysis of the Ocular Hypertension Treatment Study. Data were collected from 22 US sites with a mean (SD) follow-up of 14.0 (6.9) years. A total of 1636 participants were followed up from February 1994 to December 2008; 1077 participants were enrolled in an ancillary genetics study, of which 1009 met criteria for this analysis. PRS was calculated using summary statistics from the largest cross-ancestry POAG meta-analysis, with weights trained using 8 813 496 variants from 449 186 cross-ancestry participants in the UK Biobank. Data were analyzed from July 2022 to December 2023. Exposures From February 1994 to June 2002, participants were randomized to either topical intraocular pressure-lowering medication or close observation. After June 2002, both groups received medication. Main Outcomes and Measures Outcome measures were hazard ratios for POAG onset. Concordance index and time-dependent areas under the receiver operating characteristic curve were used to compare the predictive performance of multivariable Cox proportional hazards models. Results Of 1009 included participants, 562 (55.7%) were female, and the mean (SD) age was 55.9 (9.3) years. The mean (SD) PRS was significantly higher for 350 POAG converters (0.24 [0.95]) compared with 659 nonconverters (-0.12 [1.00]) (P < .001). POAG risk increased 1.36% (95% CI, 1.08-1.64) with each higher PRS decile, with conversion ranging from 9.52% (95% CI, 7.09-11.95) in the lowest PRS decile to 21.81% (95% CI, 19.37-24.25) in the highest decile. Comparison of low-risk and high-risk PRS tertiles showed a 2.0-fold increase in 20-year POAG risk for participants of European and African ancestries. In the subgroup randomized to delayed treatment, each increase in PRS decile was associated with a 0.52-year (95% CI, 0.01-1.03) decrease in age at diagnosis (P = .047). No significant linear association between PRS and age at POAG diagnosis was present in the early treatment group. Prediction models significantly improved with the addition of PRS as a covariate (C index = 0.77) compared with the Ocular Hypertension Treatment Study baseline model (C index = 0.75) (P < .001). Each 1-SD higher PRS conferred a mean hazard ratio of 1.25 (95% CI, 1.13-1.44) for POAG onset. Conclusions and Relevance Higher PRS was associated with increased risk for POAG in patients with ocular hypertension. The inclusion of a PRS improved the prediction of POAG onset. Trial Registration ClinicalTrials.gov Identifier: NCT00000125.
Collapse
Affiliation(s)
- Rishabh K. Singh
- Department of Ophthalmology, Columbia University Medical Center, New York, New York
- Schepens Eye Research Institute, Harvard Medical School, Boston, Massachusetts
| | - Yan Zhao
- Massachusetts Eye and Ear, Harvard Medical School, Boston
| | - Tobias Elze
- Schepens Eye Research Institute, Harvard Medical School, Boston, Massachusetts
| | - John Fingert
- Carver College of Medicine, University of Iowa, Iowa City
| | - Mae Gordon
- Washington University School of Medicine, St Louis, Missouri
| | - Michael A. Kass
- Washington University School of Medicine, St Louis, Missouri
| | - Yuyang Luo
- Massachusetts Eye and Ear, Harvard Medical School, Boston
| | | | - Todd Scheetz
- Carver College of Medicine, University of Iowa, Iowa City
| | - Ayellet V. Segrè
- Massachusetts Eye and Ear, Harvard Medical School, Boston
- Ocular Genomics Institute, Massachusetts Eye and Ear, Boston
| | - Janey L. Wiggs
- Massachusetts Eye and Ear, Harvard Medical School, Boston
- Ocular Genomics Institute, Massachusetts Eye and Ear, Boston
| | | |
Collapse
|
48
|
Grebe TA, Khushf G, Greally JM, Turley P, Foyouzi N, Rabin-Havt S, Berkman BE, Pope K, Vatta M, Kaur S. Clinical utility of polygenic risk scores for embryo selection: A points to consider statement of the American College of Medical Genetics and Genomics (ACMG). Genet Med 2024; 26:101052. [PMID: 38393332 DOI: 10.1016/j.gim.2023.101052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 02/25/2024] Open
Affiliation(s)
- Theresa A Grebe
- Phoenix Children's, Phoenix, AZ; Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ
| | - George Khushf
- Department of Philosophy, University of South Carolina, Columbia, SC
| | - John M Greally
- Departments of Genetics and Pediatrics, Albert Einstein College of Medicine, Bronx, NY
| | - Patrick Turley
- Center for Economic and Social Research, University of Southern California, Los Angeles, CA; Department of Economics, University of Southern California, Los Angeles, CA
| | | | - Sara Rabin-Havt
- Department of OB/GYN, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY
| | - Benjamin E Berkman
- Department of Bioethics, National Institutes of Health; National Human Genome Research Institute, Bethesda, MD
| | - Kathleen Pope
- Department of Pediatrics, Nemours Children's Hospital, Orlando, FL; University of South Florida College of Public Health, Tampa, FL
| | | | - Shagun Kaur
- Phoenix Children's, Phoenix, AZ; Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ
| |
Collapse
|
49
|
Liu Y, Ritchie SC, Teo SM, Ruuskanen MO, Kambur O, Zhu Q, Sanders J, Vázquez-Baeza Y, Verspoor K, Jousilahti P, Lahti L, Niiranen T, Salomaa V, Havulinna AS, Knight R, Méric G, Inouye M. Integration of polygenic and gut metagenomic risk prediction for common diseases. NATURE AGING 2024; 4:584-594. [PMID: 38528230 PMCID: PMC11031402 DOI: 10.1038/s43587-024-00590-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 02/13/2024] [Indexed: 03/27/2024]
Abstract
Multiomics has shown promise in noninvasive risk profiling and early detection of various common diseases. In the present study, in a prospective population-based cohort with ~18 years of e-health record follow-up, we investigated the incremental and combined value of genomic and gut metagenomic risk assessment compared with conventional risk factors for predicting incident coronary artery disease (CAD), type 2 diabetes (T2D), Alzheimer disease and prostate cancer. We found that polygenic risk scores (PRSs) improved prediction over conventional risk factors for all diseases. Gut microbiome scores improved predictive capacity over baseline age for CAD, T2D and prostate cancer. Integrated risk models of PRSs, gut microbiome scores and conventional risk factors achieved the highest predictive performance for all diseases studied compared with models based on conventional risk factors alone. The present study demonstrates that integrated PRSs and gut metagenomic risk models improve the predictive value over conventional risk factors for common chronic diseases.
Collapse
Affiliation(s)
- Yang Liu
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.
- Department of Clinical Pathology, Melbourne Medical School, University of Melbourne, Melbourne, Victoria, Australia.
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK.
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
| | - Scott C Ritchie
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Cambridge Centre of Research Excellence, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
| | - Shu Mei Teo
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Matti O Ruuskanen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Department of Computing, University of Turku, Turku, Finland
| | - Oleg Kambur
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Qiyun Zhu
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ, USA
| | - Jon Sanders
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, NY, USA
| | - Yoshiki Vázquez-Baeza
- Center for Microbiome Innovation, Jacobs School of Engineering, University of California San Diego, La Jolla, CA, USA
| | - Karin Verspoor
- School of Computing Technologies, RMIT University, Melbourne, Victoria, Australia
- School of Computing and Information Systems, University of Melbourne, Melbourne, Victoria, Australia
| | - Pekka Jousilahti
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Leo Lahti
- Department of Computing, University of Turku, Turku, Finland
| | - Teemu Niiranen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Division of Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Veikko Salomaa
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Aki S Havulinna
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Institute for Molecular Medicine Finland, FIMM-HiLIFE, University of Helsinki, Helsinki, Finland
| | - Rob Knight
- Center for Microbiome Innovation, Jacobs School of Engineering, University of California San Diego, La Jolla, CA, USA
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Guillaume Méric
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
- Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia
- Department of Medical Sciences, Molecular Epidemiology, Uppsala University, Uppsala, Sweden
| | - Michael Inouye
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.
- Department of Clinical Pathology, Melbourne Medical School, University of Melbourne, Melbourne, Victoria, Australia.
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK.
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- British Heart Foundation Cambridge Centre of Research Excellence, School of Clinical Medicine, University of Cambridge, Cambridge, UK.
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK.
- The Alan Turing Institute, London, UK.
| |
Collapse
|
50
|
Park YS, Jang HM, Park JH, Kim BJ, Park HY, Kim YJ. Evaluating cardiovascular disease risk stratification using multiple-polygenic risk scores and pooled cohort equations: insights from a 17-year longitudinal Korean cohort study. Front Genet 2024; 15:1364993. [PMID: 38606355 PMCID: PMC11007088 DOI: 10.3389/fgene.2024.1364993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 03/11/2024] [Indexed: 04/13/2024] Open
Abstract
Cardiovascular disease (CVD) remains the leading cause of mortality worldwide, caused by a complex interplay of genetic and environmental factors. This study aimed to evaluate the combined efficacy of multi-polygenic risk scores and pooled cohort equations (PCE) for predicting future CVD risks in the Korean population. In this longitudinal study, 7,612 individuals from the Ansan and Ansung cohorts were analyzed over a 17-year follow-up period. The participants were genotyped using the Korea Biobank Array, and quality-controlled genetic data were subjected to imputation analysis. The weighted sum of the PRSs (wPRSsum) was calculated using PRS-CS with summary statistics from myocardial infarction, ischemic stroke, coronary artery disease, and hypertension genome-wide association studies. The recalibrated PCE was used to assess clinical risk, and the participants were stratified into risk groups based on the wPRSsum and PCE. Associations between these risk scores and incident CVD were evaluated using Cox proportional hazards models and Kaplan-Meier analysis. The wPRSsum approach showed a significant association with incident CVD (HR = 1.15, p = 7.49 × 10-5), and the top 20% high-risk genetic group had an HR of 1.50 (p = 5.04 × 10-4). The recalibrated PCE effectively differentiated between the low and high 10-year CVD risk groups, with a marked difference in survival rates. The predictive models constructed using the wPRSsum and PCE demonstrated a slight improvement in prediction accuracy, particularly among males aged <55 years (C-index = 0.640). We demonstrated that while the integration of wPRSsum with PCE did not significantly outperform the PCE-only model (C-index: 0.703 for combined and 0.704 for PCE-only), it provided enhanced stratification of CVD risk. The highest risk group, identified through the combination of high wPRSsum and PCE scores, exhibited an HR of 4.99 for incident CVD (p = 1.45 × 10-15). These findings highlight the potential of integrating genetic risk assessments with traditional clinical tools for effective CVD risk stratification. Although the addition of wPRSsum to the PCE provided a marginal predictive improvement, it proved valuable in identifying high-risk individuals and supporting personalized treatment strategies. This study reinforces the utility of multi-PRS in conjunction with clinical risk assessment tools, paving the way for more tailored approaches for CVD prevention and management in diverse populations.
Collapse
Affiliation(s)
- Yi Seul Park
- Division of Genome Science, Department of Precision Medicine, National Institute of Health, Cheongju-si, Republic of Korea
| | - Hye-Mi Jang
- Division of Genome Science, Department of Precision Medicine, National Institute of Health, Cheongju-si, Republic of Korea
| | - Ji Hye Park
- Division of Genome Science, Department of Precision Medicine, National Institute of Health, Cheongju-si, Republic of Korea
| | - Bong-Jo Kim
- Division of Genome Science, Department of Precision Medicine, National Institute of Health, Cheongju-si, Republic of Korea
| | - Hyun-Young Park
- National Institute of Health, Cheongju-si, Republic of Korea
| | - Young Jin Kim
- Division of Genome Science, Department of Precision Medicine, National Institute of Health, Cheongju-si, Republic of Korea
| |
Collapse
|