1
|
Lophatananon A, Fulford G, Young J, Hart S, Brine M, Muir KR. Charity-Provided Community-Based PSA Testing for Assessment of Prostate Cancer Risk in the UK: Clinical Implications and Future Opportunities. Cancers (Basel) 2025; 17:1728. [PMID: 40427225 PMCID: PMC12109759 DOI: 10.3390/cancers17101728] [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: 11/26/2024] [Revised: 05/14/2025] [Accepted: 05/15/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Prostate cancer is the most common malignancy among UK men, yet the lack of a national screening program creates disparities in early detection. PSA testing during primary care is inconsistent, limiting timely diagnosis. The Graham Fulford Charitable Trust (GFCT) and its associated support groups offer community-based PSA testing to help bridge this gap. Objectives: This study evaluates GFCT's historical testing records and a participant survey (2021-2024) to assess their community-based PSA testing program, which is widely used and offers shared decision support. Additionally, we examine the GFCT's alignment with emerging initiatives such as the UK TRANSFORM trial and other PSA screening developments across Europe and the USA. Methods: The GFCT systematically collects PSA testing data, conducting over 50,000 tests annually. The study assesses the performance of their traffic light scoring system and a previously defined combination algorithm, "Riskman", which incorporates PSA, PSAft%, and age to classify individuals into risk categories. Multivariable logistic regression was used to evaluate and compare the predictive performance of each approach. Results: Based on the self-reported data in the survey, the GFCT's traffic light system-using age-specific PSA thresholds to assign green, amber, or red risk-showed good predictive ability. Men in the red group had over 15-fold increased odds of clinically significant cancer (Grade Group ≥ 3) compared to those in the green group. While the Riskman score achieved a higher AUC (0.84 vs. 0.76), both tools were effective in identifying high-risk individuals. Conclusions: This study highlights the GFCT's role in improving access to PSA screening and integrating practical risk stratification. Its alignment with evolving screening initiatives demonstrates the value of community-based, data-driven approaches for earlier detection of aggressive prostate cancer.
Collapse
Affiliation(s)
- Artitaya Lophatananon
- Division of Population Health, Health Services Research and Primary Care School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK;
| | - Graham Fulford
- GFCT Ltd. Formerly, The Graham Fulford Charitable Trust, 66b Smith Street, Warwick CV34 4HU, UK
| | - Jon Young
- GFCT Ltd. Formerly, The Graham Fulford Charitable Trust, 66b Smith Street, Warwick CV34 4HU, UK
| | - Susan Hart
- GFCT Ltd. Formerly, The Graham Fulford Charitable Trust, 66b Smith Street, Warwick CV34 4HU, UK
| | - Matthew Brine
- Empresa Limited, 43 All Saints Green, Norwich NR1 3LY, UK
| | - Kenneth R. Muir
- Division of Population Health, Health Services Research and Primary Care School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK;
| |
Collapse
|
2
|
Iyer HS, Cheng I, Opara C, Lin K, Zeinomar N, Le Marchand L, Wilkens L, Shariff-Marco S, Conti DV, Haiman CA, Gomez SL, Rebbeck TR. African Genetic Ancestry, Structural and Social Determinants of Health, and Mortality in Black Adults. JAMA Netw Open 2025; 8:e2510016. [PMID: 40358946 PMCID: PMC12076178 DOI: 10.1001/jamanetworkopen.2025.10016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 02/22/2025] [Indexed: 05/15/2025] Open
Abstract
Importance Although structural and social determinants of health (SSDH) have been consistently associated with health disparities, percentage African genetic ancestry (AGA) has been suggested as a risk factor associated with common diseases in Black populations. Appropriate use and interpretation of percentage AGA in understanding health disparities has been complicated by the fact that percentage AGA is correlated with genetic and nongenetic factors. Objective To evaluate associations of SSDH with mortality in the context of percentage AGA and how percentage AGA is correlated with SSDH. Design, Setting, and Participants This cohort study investigated data from the Multiethnic Cohort (MEC) Study, in which participants were enrolled from 1993 through 1996 and followed up until death or censoring on December 31, 2019. Participant data were analyzed between March and June 2023. The population-based sample was predominantly from Los Angeles County, California, consisting of self-identified Black adults aged 45 to 75 years who enrolled into the MEC Study; completed a baseline demographic, clinical, and lifestyle questionnaire; and provided biospecimens. Exposures The Index of Concentration at the Extremes (ICE), capturing social polarization based on income and racial composition, and a neighborhood socioeconomic status (NSES) index were computed from the 1990 Census, scaled to county-specific quintiles, and linked to residential census tracts at study enrollment. Percentage AGA was estimated using 21 431 single-nucleotide variations based on similarity with African continental referent data. Main Outcomes and Measures Multivariable hazard ratios (HRs) for all-cause mortality were estimated from Cox models. Correlation of percentage AGA with SSDH measures was described. Results After exclusions, 9685 participants were included (mean [SD] age, 61.0 [8.9] years; 5593 female [57.7%]), with a mean (SD) percentage AGA of 75.0% (14.0%). There were 5504 deaths over 204 463 person-years of follow-up. Comparing the most with least advantaged quintile, income ICE (adjusted HR [aHR], 1.30; 95% CI, 1.16-1.45) and NSES (aHR, 1.37, 95% CI, 1.20-1.56) were associated with lower all-cause mortality. Minimal changes were observed after adjusting for percentage AGA; for example, comparing the most with least advantaged quintile, NSES (aHR, 1.36; 95% CI, 1.19-1.55) remained associated with lower all-cause mortality. There was no association between percentage AGA and mortality after adjustment (aHR per 10-percentage point change in percentage AGA, 1.01; 95% CI, 0.99-1.03). Conclusions and Relevance In this study, associations of SSDH with mortality persisted with adjustment for percentage AGA. Findings support the hypothesis that SSDH should be the primary factors to consider for eliminating health disparities.
Collapse
Affiliation(s)
- Hari S. Iyer
- Section of Cancer Epidemiology and Health Outcomes, Rutgers Cancer Institute, New Brunswick, New Jersey
| | - Iona Cheng
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - Chidinma Opara
- Section of Cancer Epidemiology and Health Outcomes, Rutgers Cancer Institute, New Brunswick, New Jersey
| | - Katherine Lin
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - Nur Zeinomar
- Section of Cancer Epidemiology and Health Outcomes, Rutgers Cancer Institute, New Brunswick, New Jersey
| | - Loïc Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu
| | - Lynne Wilkens
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu
| | - Salma Shariff-Marco
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - David V. Conti
- Center for Genetic Epidemiology, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles
| | - Christopher A. Haiman
- Center for Genetic Epidemiology, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles
| | - Scarlett L. Gomez
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - Timothy R. Rebbeck
- Division of Population Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| |
Collapse
|
3
|
Young RP, Scott RJ, Callender T, Duan F, Billings P, Aberle DR, Gamble GD. Polygenic Risk Score Is Associated with Developing and Dying from Lung Cancer in the National Lung Screening Trial. J Clin Med 2025; 14:3110. [PMID: 40364136 PMCID: PMC12073000 DOI: 10.3390/jcm14093110] [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: 03/25/2025] [Revised: 04/17/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Background: Epidemiological studies suggest lung cancer results from the combined effects of smoking and genetic susceptibility. The clinical application of polygenic risk scores (PRSs), derived from combining the results from multiple germline genetic variants, have not yet been explored in a lung cancer screening cohort. Methods: This was a post hoc analysis of 9191 non-Hispanic white subjects from the National Lung Screening Trial (NLST), a sub-study of high-risk smokers randomised to annual computed tomography (CT) or chest X-ray (CXR) and followed for 6.4 years (mean). This study's primary aim was to examine the relationship between a composite polygenic risk score (PRS) calculated from 12 validated risk genotypes and developing or dying from lung cancer during screening. Validation was undertaken in the UK Biobank of unscreened ever-smokers (N = 167,796) followed for 10 years (median). Results: In this prospective study, we found our PRS correlated with lung cancer incidence (p < 0.0001) and mortality (p = 0.004). In an adjusted multivariable logistic regression analysis, PRS was independently associated with lung cancer death (p = 0.0027). Screening participants with intermediate and high PRS scores had a higher lung cancer mortality, relative to those with a low PRS score (rate ratios = 1.73 (95%CI 1.14-2.64, p = 0.010) and 1.89 (95%CI 1.28-2.78, p = 0.009), respectively). This was despite comparable baseline demographics (including lung function) and comparable lung cancer characteristics. The PRS's association with lung cancer mortality was validated in an unscreened cohort from the UK Biobank (p = 0.002). Conclusions: In this biomarker-based cohort study, an elevated PRS was independently associated with dying from lung cancer in both screening and non-screening cohorts.
Collapse
Affiliation(s)
- Robert P. Young
- Faculty of Medical and Health Sciences, University of Auckland, Auckland P.O. Box 37-971, New Zealand; (R.J.S.); (G.D.G.)
- Respiratory Research Group, Greenlane Clinical Centre, Epsom, Auckland 1344, New Zealand
| | - Raewyn J Scott
- Faculty of Medical and Health Sciences, University of Auckland, Auckland P.O. Box 37-971, New Zealand; (R.J.S.); (G.D.G.)
| | - Tom Callender
- Department of Applied Health Research, University College London, London WC1E6B1, UK;
| | - Fenghai Duan
- Department of Biostatistics and Centre for Biostatistics and Health Data Science, Brown University of Public Health, Providence, RI 02912, USA;
| | | | - Denise R. Aberle
- Department of Radiological Sciences, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA;
| | - Greg D. Gamble
- Faculty of Medical and Health Sciences, University of Auckland, Auckland P.O. Box 37-971, New Zealand; (R.J.S.); (G.D.G.)
| |
Collapse
|
4
|
Xu L, Zhou G, Jiang W, Zhang H, Dong Y, Guan L, Zhao H. JointPRS: A data-adaptive framework for multi-population genetic risk prediction incorporating genetic correlation. Nat Commun 2025; 16:3841. [PMID: 40268942 PMCID: PMC12019179 DOI: 10.1038/s41467-025-59243-x] [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: 11/11/2024] [Accepted: 04/16/2025] [Indexed: 04/25/2025] Open
Abstract
Genetic risk prediction for non-European populations is hindered by limited Genome-Wide Association Study (GWAS) sample sizes and small tuning datasets. We propose JointPRS, a data-adaptive framework that leverages genetic correlations across multiple populations using GWAS summary statistics. It achieves accurate predictions without individual-level tuning data and remains effective in the presence of a small tuning set thanks to its data-adaptive approach. Through extensive simulations and real data applications to 22 quantitative and four binary traits in five continental populations evaluated using the UK Biobank (UKBB) and All of Us (AoU), JointPRS consistently outperforms six state-of-the-art methods across three data scenarios: no tuning data, same-cohort tuning and testing, and cross-cohort tuning and testing. Notably, in the Admixed American population, JointPRS improves lipid trait prediction in AoU by 6.46%-172.00% compared to the other existing methods.
Collapse
Affiliation(s)
- Leqi Xu
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - Geyu Zhou
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - Wei Jiang
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
- Department of Mathematics, University of Texas at Arlington, Arlington, Texas, USA
- Division of Data Science, College of Science, University of Texas at Arlington, Arlington, Texas, USA
| | - Haoyu Zhang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Yikai Dong
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - Leying Guan
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA.
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA.
| | - Hongyu Zhao
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA.
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA.
| |
Collapse
|
5
|
Jenkins CA, De Risio L, Lophatananon A, Lewis TW, Foster D, Johnson J, Lohi H, Mellersh CS, Ricketts SL. Genome-wide association study of idiopathic epilepsy in the Italian Spinone dog breed. PLoS One 2025; 20:e0315546. [PMID: 40043055 PMCID: PMC11882058 DOI: 10.1371/journal.pone.0315546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 11/27/2024] [Indexed: 03/09/2025] Open
Abstract
Idiopathic epilepsy (IE) has a high prevalence and a severe clinical course in the Italian Spinone breed of dog. A genome-wide association study meta-analysis of 52 cases and 51 controls was conducted to identify genomic regions that may be involved with the development of IE. Subsequent to the meta-analysis, a set of 175 controls and an independent validation set of 23 cases and 23 controls were genotyped for SNPs showing suggestive association with IE to find variants exhibiting evidence of replicable association and to test the predictiveness of SNPs for IE status when combined in a weighted risk score. Although two regions showed statistically significant association with IE in the GWAS meta-analysis, and additional regions with suggestive association were identified, the findings were not emulated in the validation set. This is the first GWAS of IE in the Italian Spinone, and the findings suggest that IE in the breed is not monogenic and demonstrates the challenges when investigating a multigenic or complex inherited disease in a numerically small domesticated animal population.
Collapse
Affiliation(s)
- Christopher A. Jenkins
- Department of Veterinary Medicine, Canine Genetics Centre, University of Cambridge, Cambridge, United Kingdom (Formerly at the Animal Health Trust, Newmarket, Suffolk, United Kingdom),
- Division of Population Health, Health Services Research & Primary Care, University of Manchester, Manchester, United Kingdom
| | - Luisa De Risio
- Neurology/Neurosurgery Service, Centre for Small Animal Studies, Animal Health Trust, Newmarket, Suffolk, United Kingdom
- Linnaeus Veterinary Ltd, Shirley, Solihull, United Kingdom
| | - Artitaya Lophatananon
- Division of Population Health, Health Services Research & Primary Care, University of Manchester, Manchester, United Kingdom
| | - Thomas W. Lewis
- The Kennel Club, London, United Kingdom
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, United Kingdom
| | - Donna Foster
- Department of Veterinary Medicine, Canine Genetics Centre, University of Cambridge, Cambridge, United Kingdom (Formerly at the Animal Health Trust, Newmarket, Suffolk, United Kingdom),
| | - Jim Johnson
- Department of Veterinary Medicine, Canine Genetics Centre, University of Cambridge, Cambridge, United Kingdom (Formerly at the Animal Health Trust, Newmarket, Suffolk, United Kingdom),
| | - Hannes Lohi
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
| | - Cathryn S. Mellersh
- Department of Veterinary Medicine, Canine Genetics Centre, University of Cambridge, Cambridge, United Kingdom (Formerly at the Animal Health Trust, Newmarket, Suffolk, United Kingdom),
| | - Sally L. Ricketts
- Department of Veterinary Medicine, Canine Genetics Centre, University of Cambridge, Cambridge, United Kingdom (Formerly at the Animal Health Trust, Newmarket, Suffolk, United Kingdom),
- Division of Population Health, Health Services Research & Primary Care, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
6
|
Dornisch AM, Xu GJ, Karunamuni R, Brunette CA, Danowski ME, Teerlink CC, Gaziano JM, Garraway IP, Hauger RL, Kibel AS, Lynch JA, Maxwell KN, Rose BS, Andreassen OA, Dale AM, Donovan JL, Hamdy F, Lane A, Mills IG, Martin RM, Neal DE, Turner EL, Wolk A, Vassy JL, Seibert T. Specificity of a polygenic score for aggressive prostate cancer. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.23.25321041. [PMID: 39974097 PMCID: PMC11838993 DOI: 10.1101/2025.01.23.25321041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
To address the concern that polygenic hazard scores for prostate cancer (PCa) might not distinguish between indolent and aggressive disease, we performed case-only analyses using a 601-variant polygenic score (PHS601). We hypothesized that among men who eventually developed PCa, those with higher PHS were more likely to develop aggressive disease. We analyzed genetic and phenotypic data from a diverse, national cohort of men diagnosed with PCa (Million Veteran Program, n = 69,901, 6413 metastatic). We used Cox proportional hazards models to examine the association of PHS601 with both age at onset of metastatic PCa (birth-to-met) and time from localized to metastatic diagnosis (localized-to-met). We performed an external validation of the case-only birth-to-met analysis in two population-based cohorts from the PRACTICAL Consortium. PHS601 was associated with both birth-to-met and localized-to-met within MVP. The HRs for men in the highest quintile of PHS601 vs. those in the lowest quintile (HR 80/20 ) were 1.74 [1.65-1.84] and 1.41 [1.31-1.41] for birth-to-met and localized-to-met, respectively. These findings were validated in two external cohorts (COSM and ProtecT). PHS601 was also associated with earlier development of metastasis. Men with high PHS601 are diagnosed with prostate cancer at a younger age and are more likely to develop metastasis.
Collapse
|
7
|
Kaushal JB, Raut P, Muniyan S, Siddiqui JA, Alsafwani ZW, Seshacharyulu P, Nair SS, Tewari AK, Batra SK. Racial disparity in prostate cancer: an outlook in genetic and molecular landscape. Cancer Metastasis Rev 2024; 43:1233-1255. [PMID: 38902476 PMCID: PMC11560487 DOI: 10.1007/s10555-024-10193-8] [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: 11/27/2023] [Accepted: 06/04/2024] [Indexed: 06/22/2024]
Abstract
Prostate cancer (PCa) incidence, morbidity, and mortality rates are significantly impacted by racial disparities. Despite innovative therapeutic approaches and advancements in prevention, men of African American (AA) ancestry are at a higher risk of developing PCa and have a more aggressive and metastatic form of the disease at the time of initial PCa diagnosis than other races. Research on PCa has underlined the biological and molecular basis of racial disparity and emphasized the genetic aspect as the fundamental component of racial inequality. Furthermore, the lower enrollment rate, limited access to national-level cancer facilities, and deferred treatment of AA men and other minorities are hurdles in improving the outcomes of PCa patients. This review provides the most up-to-date information on various biological and molecular contributing factors, such as the single nucleotide polymorphisms (SNPs), mutational spectrum, altered chromosomal loci, differential gene expression, transcriptome analysis, epigenetic factors, tumor microenvironment (TME), and immune modulation of PCa racial disparities. This review also highlights future research avenues to explore the underlying biological factors contributing to PCa disparities, particularly in men of African ancestry.
Collapse
Affiliation(s)
- Jyoti B Kaushal
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE-68198, USA
| | - Pratima Raut
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE-68198, USA
| | - Sakthivel Muniyan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE-68198, USA
| | - Jawed A Siddiqui
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE-68198, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE-68198, USA
| | - Zahraa W Alsafwani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE-68198, USA
| | - Parthasarathy Seshacharyulu
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE-68198, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE-68198, USA
| | - Sujit S Nair
- Department of Urology and the Tisch Cancer Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ashutosh K Tewari
- Department of Urology and the Tisch Cancer Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE-68198, USA.
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE-68198, USA.
- Division of Urology, Department of Surgery, University of Nebraska Medical Center, Omaha, NE-68198, USA.
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE-68198, USA.
| |
Collapse
|
8
|
Lippert AM, Corsi DJ, Kim R, Wedow R, Kim J, Taddess B, Subramanian SV. Polygenic and Socioeconomic Contributions to Nicotine Use and Cardiometabolic Health in Early Mid-Life. Nicotine Tob Res 2024; 26:1616-1625. [PMID: 38874009 DOI: 10.1093/ntr/ntae146] [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: 09/21/2023] [Revised: 05/18/2024] [Accepted: 06/07/2024] [Indexed: 06/15/2024]
Abstract
INTRODUCTION Early mid-life is marked by accumulating risks for cardiometabolic illness linked to health-risk behaviors like nicotine use. Identifying polygenic indices (PGI) has enriched scientific understanding of the cumulative genetic contributions to behavioral and cardiometabolic health, though few studies have assessed these associations alongside socioeconomic (SES) and lifestyle factors. AIMS AND METHODS Drawing on data from 2337 individuals from the United States participating in the National Longitudinal Study of Adolescent to Adult Health, the current study assesses the fraction of variance in five related outcomes-use of conventional and electronic cigarettes, body mass index (BMI), waist circumference, and glycosylated hemoglobin (A1c)-explained by PGI, SES, and lifestyle. RESULTS Regression models on African ancestry (AA) and European ancestry (EA) subsamples reveal that the fraction of variance explained by PGI ranges across outcomes. While adjusting for sex and age, PGI explained 3.5%, 2.2%, and 0% in the AA subsample of variability in BMI, waist circumference, and A1c, respectively (in the EA subsample these figures were 7.7%, 9.4%, and 1.3%). The proportion of variance explained by PGI in nicotine-use outcomes is also variable. Results further indicate that PGI and SES are generally complementary, accounting for more variance in the outcomes when modeled together versus separately. CONCLUSIONS PGI are gaining attention in population health surveillance, but polygenic variability might not align clearly with health differences in populations or surpass SES as a fundamental cause of health disparities. We discuss future steps in integrating PGI and SES to refine population health prediction rules. IMPLICATIONS Study findings point to the complementary relationship of PGI and socioeconomic indicators in explaining population variance in nicotine outcomes and cardiometabolic wellness. Population health surveillance and prediction rules would benefit from the combination of information from both polygenic and socioeconomic risks. Additionally, the risk for electronic cigarette use among users of conventional cigarettes may have a genetic component tied to the cumulative genetic propensity for heavy smoking. Further research on PGI for vaping is needed.
Collapse
Affiliation(s)
- Adam M Lippert
- Sociology Department, University of Colorado Denver, Denver, CO, USA
| | - Daniel J Corsi
- Ottawa Hospital Research Institute, Clinical Epidemiology Program, Ottawa, ON, Canada
| | - Rockli Kim
- Division of Health Policy and Management, College of Health Science, Korea University, Seoul, South Korea
- Interdisciplinary Program in Precision Public Health, Department of Public Health Sciences, Graduate School of Korea University, Seoul, South Korea
| | - Robbee Wedow
- Sociology Department, Purdue University, West Layfette, IN, USA
| | - Jinho Kim
- Division of Health Policy and Management, College of Health Science, Korea University, Seoul, South Korea
- Interdisciplinary Program in Precision Public Health, Department of Public Health Sciences, Graduate School of Korea University, Seoul, South Korea
| | - Beza Taddess
- Sociology Department, Princeton University, Princeton, NJ, USA
| | - S V Subramanian
- Center for Population and Development Studies, Harvard University, Cambridge, MA, USA
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
9
|
Spears C, Xu M, Shoben A, Dason S, Toland AE, Byrne L. Clinical features of prostate cancer by polygenic risk score. Fam Cancer 2024; 23:499-505. [PMID: 38619781 PMCID: PMC11512885 DOI: 10.1007/s10689-024-00369-0] [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: 11/09/2023] [Accepted: 02/25/2024] [Indexed: 04/16/2024]
Abstract
Genome-wide association studies have identified more than 290 single nucleotide variants (SNVs) associated with prostate cancer. These SNVs can be combined to generate a Polygenic Risk Score (PRS), which estimates an individual's risk to develop prostate cancer. Identifying individuals at higher risk for prostate cancer using PRS could allow for personalized screening recommendations, improve current screening tools, and potentially result in improved survival rates, but more research is needed before incorporating them into clinical use. Our study aimed to investigate associations between PRS and clinical factors in affected individuals, including age of diagnosis, metastases, histology, International Society of Urological Pathology (ISUP) Grade Group (GG) and family history of prostate cancer, while taking into account germline genetic testing in known prostate cancer related genes. To evaluate the relationship between these clinical factors and PRS, a quantitative retrospective chart review of 250 individuals of European ancestry diagnosed with prostate cancer who received genetic counseling services at The Ohio State University's Genitourinary Cancer Genetics Clinic and a 72-SNV PRS through Ambry Genetics, was performed. We found significant associations between higher PRS and younger age of diagnosis (p = 0.002), lower frequency of metastases (p = 0.006), and having a first-degree relative diagnosed with prostate cancer (p = 0.024). We did not observe significant associations between PRS and ISUP GG, histology or a having a second-degree relative with prostate cancer. These findings provide insights into features associated with higher PRS, but larger multi-ancestral studies using PRS that are informative across populations are needed to understand its clinical utility.
Collapse
Affiliation(s)
- Christina Spears
- Division of Human Genetics, Department of Internal Medicine, College of Medicine, The Ohio State University, 2012 Kenny Road, Columbus, OH, 43212, USA.
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| | - Menglin Xu
- Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Abigail Shoben
- Division of Biostatistics, College of Public Health, The Ohio State University, Columbus, OH, USA
| | - Shawn Dason
- Division of Urologic Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Amanda Ewart Toland
- Division of Human Genetics, Department of Internal Medicine, College of Medicine, The Ohio State University, 2012 Kenny Road, Columbus, OH, 43212, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Lindsey Byrne
- Division of Human Genetics, Department of Internal Medicine, College of Medicine, The Ohio State University, 2012 Kenny Road, Columbus, OH, 43212, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
10
|
Pagadala MS, Lui A, Lynch J, Karunamuni R, Lee KM, Plym A, Rose BS, Carter H, Kibel AS, DuVall SL, Vassy J, Gaziano JM, Panizzon MS, Hauger RL, Seibert TM. Healthy lifestyle and prostate cancer risk in the Million Veteran Program. Cancer 2024; 130:3496-3505. [PMID: 38865417 DOI: 10.1002/cncr.35434] [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: 01/27/2024] [Revised: 04/24/2024] [Accepted: 05/21/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND This study aims to assess the impact of healthy lifestyle on prostate cancer (PCa) risk in a diverse population. METHODS Data for 281,923 men from the Million Veteran Program (MVP), a nationwide, health system-based cohort study, were analyzed. Self-reported information at enrollment included smoking status, exercise, diet, family history of PCa, and race/ethnicity. Body mass index (BMI) was obtained from clinical records. Genetic risk was assessed via a validated polygenic score. Cox proportional hazards models were used to assess associations with PCa outcomes. RESULTS After accounting for ancestry, family history, and genetic risk, smoking was associated with an increased risk of metastatic PCa (hazard ratio [HR], 1.83; 95% confidence interval [CI], 1.64-2.02; p < 10-16) and fatal PCa (HR, 2.73; 95% CI, 2.36-3.25; p < 10-16). Exercise was associated with a reduced risk of fatal PCa (HR, 0.86; 95% CI, 0.76-0.98; p = .03). Higher BMI was associated with a slightly reduced risk of fatal PCa, and diet score was not independently associated with any end point. Association with exercise was strongest among those who had nonmetastatic PCa at MVP enrollment. Absolute reductions in the risk of fatal PCa via lifestyle factors were greatest among men of African ancestry (1.7% for nonsmokers vs. 6.1% for smokers) or high genetic risk (1.4% for nonsmokers vs. 4.3% for smokers). CONCLUSIONS Healthy lifestyle is minimally related to the overall risk of developing PCa but is associated with a substantially reduced risk of dying from PCa. In multivariable analyses, both exercise and not smoking remain independently associated with reduced metastatic and fatal PCa.
Collapse
Affiliation(s)
- Meghana S Pagadala
- Research Service, Veterans Affairs (VA) San Diego Healthcare System, San Diego, California, USA
- Medical Scientist Training Program, University of California San Diego, La Jolla, California, USA
- Biomedical Science Program, University of California San Diego, La Jolla, California, USA
| | - Asona Lui
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California, USA
- Cancer Center, The Salk Institute for Biological Studies, La Jolla, California, USA
| | - Julie Lynch
- VA Informatics and Computing Infrastructure, VA Salt Lake City Healthcare System, Salt Lake City, Utah, USA
- Division of Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Roshan Karunamuni
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California, USA
| | - Kyung Min Lee
- VA Informatics and Computing Infrastructure, VA Salt Lake City Healthcare System, Salt Lake City, Utah, USA
| | - Anna Plym
- Department of Urology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Epidemiology, T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Brent S Rose
- Research Service, Veterans Affairs (VA) San Diego Healthcare System, San Diego, California, USA
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California, USA
| | - Hannah Carter
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Adam S Kibel
- Department of Urology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Scott L DuVall
- VA Informatics and Computing Infrastructure, VA Salt Lake City Healthcare System, Salt Lake City, Utah, USA
- Division of Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Jason Vassy
- VA Boston Healthcare System, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - J Michael Gaziano
- Massachusetts Veterans Epidemiology Research and Information Center, VA Boston Healthcare System, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Matthew S Panizzon
- Research Service, Veterans Affairs (VA) San Diego Healthcare System, San Diego, California, USA
- Center for Behavioral Genetics of Aging, University of California San Diego, La Jolla, California, USA
| | - Richard L Hauger
- Research Service, Veterans Affairs (VA) San Diego Healthcare System, San Diego, California, USA
- Center for Behavioral Genetics of Aging, University of California San Diego, La Jolla, California, USA
- Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, San Diego, California, USA
| | - Tyler M Seibert
- Research Service, Veterans Affairs (VA) San Diego Healthcare System, San Diego, California, USA
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California, USA
- Department of Radiology, University of California San Diego, La Jolla, California, USA
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
11
|
Lee PH, Chen IC, Chen YM, Hsiao TH, Tseng JS, Huang YH, Hsu KH, Lin H, Yang TY, Shao YHJ. Using a Polygenic Risk Score to Improve the Risk Prediction of Non-Small Cell Lung Cancer in Taiwan. JCO Precis Oncol 2024; 8:e2400236. [PMID: 39348659 DOI: 10.1200/po.24.00236] [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: 04/08/2024] [Revised: 07/18/2024] [Accepted: 08/20/2024] [Indexed: 10/02/2024] Open
Abstract
PURPOSE Low-dose computed tomography (LDCT) can help reducing lung cancer mortality. In Taiwan, the existing screening criteria revolve around smoking habits and family history of lung cancer. The role of genetic variation in non-small cell lung cancer (NSCLC) development is increasingly recognized. In this study, we aimed to investigate the potential benefits of polygenic risk scores (PRSs) in predicting NSCLC and enhancing the effectiveness of screening programs. METHODS We conducted a retrospective cohort study that included participants without prior diagnosis of lung cancer and later received LDCT for lung cancer screening. Genetic data for these participants were obtained from the project of Taiwan Precision Medicine Initiative. We adopted the model of genome-wide association study-derived PRS calculation using 19 susceptibility loci associated with the risk of NSCLC as reported by Dai et al. RESULTS We studied a total of 2,287 participants (1,197 male, 1,090 female). More female participants developed NSCLC during the follow-up period (4.4% v 2.5%, P = .015). The only risk factor of NSCLC diagnosis among male participants was age. Among female participants, independent risk factors of NSCLC diagnosis were age (adjusted hazard ratio [aHR], 1.08 [95% CI, 1.04 to 1.11]), a family history of lung cancer (aHR, 3.21 [95% CI, 1.78 to 5.77]), and PRS fourth quartile (aHR, 2.97 [95% CI, 1.25 to 7.07]). We used the receiver operating characteristics to show an AUC value of 0.741 for the conventional model. With the further incorporation of PRS, the AUC rose to 0.778. CONCLUSION The evaluation of PRS for NSCLC prediction holds promise for enhancing the effectiveness of lung cancer screening in Taiwan especially in women. By incorporating genetic information, screening criteria can be tailored to identify individuals at higher risks of NSCLC.
Collapse
Affiliation(s)
- Po-Hsin Lee
- Division of Chest Medicine, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Doctoral Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan
- Rong Hsing Translational Medicine Research Center, National Chung Hsing University, Taichung, Taiwan
| | - I-Chieh Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yi-Ming Chen
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Rong Hsing Translational Medicine Research Center, National Chung Hsing University, Taichung, Taiwan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
- Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Tzu-Hung Hsiao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Public Health, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
- Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung, Taiwan
| | - Jeng-Sen Tseng
- Division of Chest Medicine, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Yen-Hsiang Huang
- Division of Chest Medicine, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Kuo-Hsuan Hsu
- Division of Critical Care and Respiratory Therapy, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Ho Lin
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Tsung-Ying Yang
- Division of Chest Medicine, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Yu-Hsuan Joni Shao
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Clinical Big Data Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| |
Collapse
|
12
|
Hall R, Bancroft E, Pashayan N, Kote-Jarai Z, Eeles RA. Genetics of prostate cancer: a review of latest evidence. J Med Genet 2024; 61:915-926. [PMID: 39137963 DOI: 10.1136/jmg-2024-109845] [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: 01/02/2024] [Accepted: 07/04/2024] [Indexed: 08/15/2024]
Abstract
Prostate cancer (PrCa) is a largely heritable and polygenic disease. It is the most common cancer in people with prostates (PwPs) in Europe and the USA, including in PwPs of African descent. In the UK in 2020, 52% of all cancers were diagnosed at stage I or II. The National Health Service (NHS) long-term plan is to increase this to 75% by 2028, to reduce absolute incidence of late-stage disease. In the absence of a UK PrCa screening programme, we should explore how to identify those at increased risk of clinically significant PrCa.Incorporating genomics into the PrCa screening, diagnostic and treatment pathway has huge potential for transforming patient care. Genomics can increase efficiency of PrCa screening by focusing on those with genetic predisposition to cancer-which when combined with risk factors such as age and ethnicity, can be used for risk stratification in risk-based screening (RBS) programmes. The goal of RBS is to facilitate early diagnosis of clinically significant PrCa and reduce overdiagnosis/overtreatment in those unlikely to experience PrCa-related symptoms in their lifetime. Genetic testing can guide PrCa management, by identifying those at risk of lethal PrCa and enabling access to novel targeted therapies.PrCa is curable if diagnosed below stage III when most people do not experience symptoms. RBS using genetic profiling could be key here if we could show better survival outcomes (or reduction in cancer-specific mortality accounting for lead-time bias), in addition to more cost efficiency than age-based screening alone. Furthermore, PrCa outcomes in underserved communities could be optimised if genetic testing was accessible, minimising health disparities.
Collapse
Affiliation(s)
- Rose Hall
- The Royal Marsden NHS Foundation Trust, London, UK
- Institute for Cancer Research, London, UK
| | | | | | | | - Rosalind A Eeles
- The Royal Marsden NHS Foundation Trust, London, UK
- Institute for Cancer Research, London, UK
| |
Collapse
|
13
|
Aznar-Gimeno R, García-González MA, Muñoz-Sierra R, Carrera-Lasfuentes P, Rodrigálvarez-Chamarro MDLV, González-Muñoz C, Meléndez-Estrada E, Lanas Á, Del Hoyo-Alonso R. GastricAITool: A Clinical Decision Support Tool for the Diagnosis and Prognosis of Gastric Cancer. Biomedicines 2024; 12:2162. [PMID: 39335675 PMCID: PMC11429470 DOI: 10.3390/biomedicines12092162] [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: 08/11/2024] [Revised: 09/10/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND/OBJECTIVE Gastric cancer (GC) is a complex disease representing a significant global health concern. Advanced tools for the early diagnosis and prediction of adverse outcomes are crucial. In this context, artificial intelligence (AI) plays a fundamental role. The aim of this work was to develop a diagnostic and prognostic tool for GC, providing support to clinicians in critical decision-making and enabling personalised strategies. METHODS Different machine learning and deep learning techniques were explored to build diagnostic and prognostic models, ensuring model interpretability and transparency through explainable AI methods. These models were developed and cross-validated using data from 590 Spanish Caucasian patients with primary GC and 633 cancer-free individuals. Up to 261 variables were analysed, including demographic, environmental, clinical, tumoral, and genetic data. Variables such as Helicobacter pylori infection, tobacco use, family history of GC, TNM staging, metastasis, tumour location, treatment received, gender, age, and genetic factors (single nucleotide polymorphisms) were selected as inputs due to their association with the risk and progression of the disease. RESULTS The XGBoost algorithm (version 1.7.4) achieved the best performance for diagnosis, with an AUC value of 0.68 using 5-fold cross-validation. As for prognosis, the Random Survival Forest algorithm achieved a C-index of 0.77. Of interest, the incorporation of genetic data into the clinical-demographics models significantly increased discriminatory ability in both diagnostic and prognostic models. CONCLUSIONS This article presents GastricAITool, a simple and intuitive decision support tool for the diagnosis and prognosis of GC.
Collapse
Affiliation(s)
- Rocío Aznar-Gimeno
- Department of Big Data and Cognitive Systems, Instituto Tecnológico de Aragón, ITA, María de Luna 7-8, 50018 Zaragoza, Spain
| | - María Asunción García-González
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain
- Instituto Aragonés de Ciencias de la Salud (IACS), 50009 Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
| | - Rubén Muñoz-Sierra
- Department of Big Data and Cognitive Systems, Instituto Tecnológico de Aragón, ITA, María de Luna 7-8, 50018 Zaragoza, Spain
| | - Patricia Carrera-Lasfuentes
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain
- Facultad de Ciencias de la Salud, Universidad San Jorge, 50830 Zaragoza, Spain
| | | | - Carlos González-Muñoz
- Department of Big Data and Cognitive Systems, Instituto Tecnológico de Aragón, ITA, María de Luna 7-8, 50018 Zaragoza, Spain
| | - Enrique Meléndez-Estrada
- Department of Big Data and Cognitive Systems, Instituto Tecnológico de Aragón, ITA, María de Luna 7-8, 50018 Zaragoza, Spain
| | - Ángel Lanas
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
- Department of Gastroenterology, Hospital Clínico Universitario Lozano Blesa, 50009 Zaragoza, Spain
- School of Medicine, University of Zaragoza, 50009 Zaragoza, Spain
| | - Rafael Del Hoyo-Alonso
- Department of Big Data and Cognitive Systems, Instituto Tecnológico de Aragón, ITA, María de Luna 7-8, 50018 Zaragoza, Spain
| |
Collapse
|
14
|
Hung SC, Chang LW, Hsiao TH, Wei CY, Wang SS, Li JR, Chen IC. Predictive value of polygenic risk score for prostate cancer incidence and prognosis in the Han Chinese. Sci Rep 2024; 14:20453. [PMID: 39227454 PMCID: PMC11372043 DOI: 10.1038/s41598-024-71544-7] [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: 09/19/2023] [Accepted: 08/28/2024] [Indexed: 09/05/2024] Open
Abstract
Although prostate cancer is a common occurrence among males, the relationship between existing risk prediction models remains unclear. The objective of this hospital-based retrospective study is to investigate the impact of polygenic risk scores (PRSs) on the incidence and prognosis of prostate cancer in the Han Chinese population. A total of 24,778 male participants including 903 patients with prostate cancer at Taichung Veterans General Hospital were enrolled in the study. PRS was calculated using 269 single nucleotide polymorphisms and their corresponding effect sizes from the polygenic score catalog. The association between PRS and the risk prostate cancer was evaluated using Cox proportional hazards regression model. Among the 24,778 participants, 903 were diagnosed with prostate cancer. The risk of prostate cancer was significantly higher in the highest quartile of PRS distribution compared to the lowest (hazard ratio = 4.770, 95% CI = 3.999-5.689, p < 0.0001), with statistical significance across all age groups. Patients in the highest quartile were diagnosed with prostate cancer at a younger age (66.8 ± 8.3 vs. 69.5 ± 8.8, p = 0.002). Subgroup analysis of patients with localized or stage 4 prostate cancer showed no significant differences in biochemical failure or overall survival. This hospital-based cohort study observed that a higher PRS was associated with increased susceptibility to prostate cancer and younger age of diagnosis. However, PRS was not found to be a significant predictor of disease stage and prognosis. These findings suggest that PRS could serve as a useful tool in prostate cancer risk assessment.
Collapse
Affiliation(s)
- Sheng-Chun Hung
- Department of Urology, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Li-Wen Chang
- Department of Urology, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Tzu-Hung Hsiao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Public Health, Fu Jen Catholic University, New Taipei City, Taiwan
- Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung, Taiwan
| | - Chia-Yi Wei
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Shian-Shiang Wang
- Department of Urology, Taichung Veterans General Hospital, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
| | - Jian-Ri Li
- Department of Urology, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medicine and Nursing, Hungkuang University, Taichung, Taiwan
| | - I-Chieh Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan.
| |
Collapse
|
15
|
Sun C, Cheng X, Xu J, Chen H, Tao J, Dong Y, Wei S, Chen R, Meng X, Ma Y, Tian H, Guo X, Bi S, Zhang C, Kang J, Zhang M, Lv H, Shang Z, Lv W, Zhang R, Jiang Y. A review of disease risk prediction methods and applications in the omics era. Proteomics 2024; 24:e2300359. [PMID: 38522029 DOI: 10.1002/pmic.202300359] [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: 09/15/2023] [Revised: 03/08/2024] [Accepted: 03/12/2024] [Indexed: 03/25/2024]
Abstract
Risk prediction and disease prevention are the innovative care challenges of the 21st century. Apart from freeing the individual from the pain of disease, it will lead to low medical costs for society. Until very recently, risk assessments have ushered in a new era with the emergence of omics technologies, including genomics, transcriptomics, epigenomics, proteomics, and so on, which potentially advance the ability of biomarkers to aid prediction models. While risk prediction has achieved great success, there are still some challenges and limitations. We reviewed the general process of omics-based disease risk model construction and the applications in four typical diseases. Meanwhile, we highlighted the problems in current studies and explored the potential opportunities and challenges for future clinical practice.
Collapse
Affiliation(s)
- Chen Sun
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The EWAS Project, Harbin, China
| | - Xiangshu Cheng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The EWAS Project, Harbin, China
| | - Jing Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The EWAS Project, Harbin, China
| | - Haiyan Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Junxian Tao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The EWAS Project, Harbin, China
| | - Yu Dong
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The EWAS Project, Harbin, China
| | - Siyu Wei
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The EWAS Project, Harbin, China
| | - Rui Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Xin Meng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yingnan Ma
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The EWAS Project, Harbin, China
| | - Hongsheng Tian
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Xuying Guo
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Shuo Bi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Chen Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Jingxuan Kang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Mingming Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Hongchao Lv
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Zhenwei Shang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Wenhua Lv
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Ruijie Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yongshuai Jiang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The EWAS Project, Harbin, China
| |
Collapse
|
16
|
Liu X, Duan H, Liu S, Zhang Y, Ji Y, Zhang Y, Feng Z, Li J, Liu Y, Gao Y, Wang X, Zhang Q, Yang L, Dai H, Lyu Z, Song F, Song F, Huang Y. Preliminary effects of risk-adapted PSA screening for prostate cancer after integrating PRS-specific and age-specific variation. Front Genet 2024; 15:1387588. [PMID: 39149591 PMCID: PMC11324495 DOI: 10.3389/fgene.2024.1387588] [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: 02/18/2024] [Accepted: 07/15/2024] [Indexed: 08/17/2024] Open
Abstract
Background Although the risk of prostate cancer (PCa) varies across different ages and genetic risks, it's unclear about the effects of genetic-specific and age-specific prostate-specific antigen (PSA) screening for PCa. Methods Weighed and unweighted polygenic risk scores (PRS) were constructed to classify the participants from the PLCO trial into low- or high-PRS groups. The age-specific and PRS-specific cut-off values of PSA for PCa screening were determined with time-dependent receiver-operating-characteristic curves and area-under-curves (tdAUCs). Improved screening strategies integrating PRS-specific and age-specific cut-off values of PSA were compared to traditional PSA screening on accuracy, detection rates of high-grade PCa (Gleason score ≥7), and false positive rate. Results Weighted PRS with 80 SNPs significantly associated with PCa was determined as the optimal PRS, with an AUC of 0.631. After stratifying by PRS, the tdAUCs of PSA with a 10-year risk of PCa were 0.818 and 0.816 for low- and high-PRS groups, whereas the cut-off values were 1.42 and 1.62 ng/mL, respectively. After further stratifying by age, the age-specific cut-off values of PSA were relatively lower for low PRS (1.42, 1.65, 1.60, and 2.24 ng/mL for aged <60, 60-64, 65-69, and ≥70 years) than high PRS (1.48, 1.47, 1.89, and 2.72 ng/mL). Further analyses showed an obvious interaction of positive PSA and high PRS on PCa incidence and mortality. Very small difference in PCa risk were observed among subgroups with PSA (-) across different age and PRS, and PCa incidence and mortality with PSA (+) significantly increased as age and PRS, with highest risk for high-PRS/PSA (+) in participants aged ≥70 years [HRs (95%CI): 16.00 (12.62-20.29) and 19.48 (9.26-40.96)]. The recommended screening strategy reduced 12.8% of missed PCa, ensured high specificity, but not caused excessive false positives than traditional PSA screening. Conclusion Risk-adapted screening integrating PRS-specific and age-specific cut-off values of PSA would be more effective than traditional PSA screening.
Collapse
Affiliation(s)
- Xiaomin Liu
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology (Tianjin), National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Hongyuan Duan
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology (Tianjin), National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Siwen Liu
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology (Tianjin), National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yunmeng Zhang
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology (Tianjin), National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yuting Ji
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology (Tianjin), National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yacong Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Zhuowei Feng
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology (Tianjin), National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jingjing Li
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology (Tianjin), National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Ya Liu
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology (Tianjin), National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Ying Gao
- Health Management Center, Tianjin Medical University General Hospital, Tianjin, China
| | - Xing Wang
- Health Management Center, Tianjin Medical University General Hospital, Tianjin, China
| | - Qing Zhang
- Health Management Center, Tianjin Medical University General Hospital, Tianjin, China
| | - Lei Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Beijing Office for Cancer Prevention and Control, Peking University Cancer Hospital and Institute, Beijing, China
| | - Hongji Dai
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology (Tianjin), National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Zhangyan Lyu
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology (Tianjin), National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Fangfang Song
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology (Tianjin), National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Fengju Song
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology (Tianjin), National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yubei Huang
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology (Tianjin), National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
17
|
Lee KM, Nelson TJ, Bryant A, Teerlink CC, Gulati R, Pagadala MS, Tcheandjieu C, Pridgen KM, DuVall SL, Yamoah K, Vassy JL, Seibert TM, Hauger RL, Rose BS, Lynch JA. Genetic risk and likelihood of prostate cancer detection on first biopsy by ancestry. J Natl Cancer Inst 2024; 116:753-757. [PMID: 38212986 PMCID: PMC11077300 DOI: 10.1093/jnci/djae002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 11/03/2023] [Accepted: 12/23/2023] [Indexed: 01/13/2024] Open
Abstract
Despite differences in prostate cancer risk across ancestry groups, relative performance of prostate cancer genetic risks scores (GRS) for positive biopsy prediction in different ancestry groups is unknown. This cross-sectional retrospective analysis examines the association between a polygenic hazard score (PHS290) and risk of prostate cancer diagnosis upon first biopsy in male veterans using 2-sided tests. Our analysis included 36 717 veterans (10 297 of African ancestry). Unadjusted rates of positive first prostate biopsy increased with higher genetic risk (low risk: 34%, high risk: 58%; P < .001). Among men of African ancestry, higher genetic risk was associated with increased prostate cancer detection on first biopsy (odds ratio = 2.18, 95% confidence interval = 1.93 to 2.47), but the effect was stronger among men of European descent (odds ratio = 3.89, 95% confidence interval = 3.62 to 4.18). These findings suggest that incorporating genetic risk into prediction models could better personalize biopsy decisions, although further study is needed to achieve equitable genetic risk stratification among ancestry groups.
Collapse
Affiliation(s)
- Kyung Min Lee
- VA Informatics and Computing Infrastructure, VA Salt Lake City Health Care System, Salt Lake City, UT, USA
| | - Tyler J Nelson
- VA Informatics and Computing Infrastructure, VA Salt Lake City Health Care System, Salt Lake City, UT, USA
| | - Alex Bryant
- Department of Radiation Oncology, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI, USA
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Craig C Teerlink
- VA Informatics and Computing Infrastructure, VA Salt Lake City Health Care System, Salt Lake City, UT, USA
- Department of Internal Medicine, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Roman Gulati
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Meghana S Pagadala
- VA San Diego Healthcare System, San Diego, CA, USA
- Medical Scientist Training Program, University of California San Diego, La Jolla, CA, USA
- Biomedical Science Program, University of California San Diego, La Jolla, CA, USA
| | - Catherine Tcheandjieu
- VA Palo Alto Health Care System, Palo Alto, CA, USA
- Gladstone Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Kathryn M Pridgen
- VA Informatics and Computing Infrastructure, VA Salt Lake City Health Care System, Salt Lake City, UT, USA
- Department of Internal Medicine, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Scott L DuVall
- VA Informatics and Computing Infrastructure, VA Salt Lake City Health Care System, Salt Lake City, UT, USA
- Department of Internal Medicine, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Kosj Yamoah
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
- James A. Haley Veterans’ Hospital, Tampa, FL, USA
| | - Jason L Vassy
- Section of General Internal Medicine, VA Boston Healthcare System, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Tyler M Seibert
- VA San Diego Healthcare System, San Diego, CA, USA
- Department of Radiation Medicine and Applied Sciences, University of California, San Diego, La Jolla, CA, USA
- Department of Radiology, University of California San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Richard L Hauger
- VA San Diego Healthcare System, San Diego, CA, USA
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Brent S Rose
- VA San Diego Healthcare System, San Diego, CA, USA
- Department of Radiation Medicine and Applied Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Julie A Lynch
- VA Informatics and Computing Infrastructure, VA Salt Lake City Health Care System, Salt Lake City, UT, USA
- Department of Internal Medicine, School of Medicine, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
18
|
Cheng Y, Wu L, Xin J, Ben S, Chen S, Li H, Zhao L, Wang M, Cheng G, Du M. An early-onset specific polygenic risk score optimizes age-based risk estimate and stratification of prostate cancer: population-based cohort study. J Transl Med 2024; 22:366. [PMID: 38632662 PMCID: PMC11025178 DOI: 10.1186/s12967-024-05190-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] [Received: 01/04/2024] [Accepted: 04/11/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Early-onset prostate cancer (EOPC, ≤ 55 years) has a unique clinical entity harboring high genetic risk, but the majority of EOPC patients still substantial opportunity to be early-detected thus suffering an unfavorable prognosis. A refined understanding of age-based polygenic risk score (PRS) for prostate cancer (PCa) would be essential for personalized risk stratification. METHODS We included 167,517 male participants [4882 cases including 205 EOPC and 4677 late-onset PCa (LOPC)] from UK Biobank. A General-, an EOPC- and an LOPC-PRS were derived from age-specific genome-wide association studies. Weighted Cox proportional hazard models were applied to estimate the risk of PCa associated with PRSs. The discriminatory capability of PRSs were validated using time-dependent receiver operating characteristic (ROC) curves with additional 4238 males from PLCO and TCGA. Phenome-wide association studies underlying Mendelian Randomization were conducted to discover EOPC linking phenotypes. RESULTS The 269-PRS calculated via well-established risk variants was more strongly associated with risk of EOPC [hazard ratio (HR) = 2.35, 95% confidence interval (CI) 1.99-2.78] than LOPC (HR = 1.95, 95% CI 1.89-2.01; I2 = 79%). EOPC-PRS was dramatically related to EOPC risk (HR = 4.70, 95% CI 3.98-5.54) but not to LOPC (HR = 0.98, 95% CI 0.96-1.01), while LOPC-PRS had similar risk estimates for EOPC and LOPC (I2 = 0%). Particularly, EOPC-PRS performed optimal discriminatory capability for EOPC (area under the ROC = 0.613). Among the phenomic factors to PCa deposited in the platform of ProAP (Prostate cancer Age-based PheWAS; https://mulongdu.shinyapps.io/proap ), EOPC was preferentially associated with PCa family history while LOPC was prone to environmental and lifestyles exposures. CONCLUSIONS This study comprehensively profiled the distinct genetic and phenotypic architecture of EOPC. The EOPC-PRS may optimize risk estimate of PCa in young males, particularly those without family history, thus providing guidance for precision population stratification.
Collapse
Affiliation(s)
- Yifei Cheng
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Department of Environmental Genomics, School of Public Health, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
- The Key Laboratory of Modern Toxicology of Ministry of Education, Department of Genetic Toxicology, School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Lang Wu
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Junyi Xin
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Shuai Ben
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Department of Environmental Genomics, School of Public Health, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
- The Key Laboratory of Modern Toxicology of Ministry of Education, Department of Genetic Toxicology, School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Silu Chen
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Department of Environmental Genomics, School of Public Health, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
- The Key Laboratory of Modern Toxicology of Ministry of Education, Department of Genetic Toxicology, School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Huiqin Li
- Department of Biostatistics, School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Lingyan Zhao
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Department of Environmental Genomics, School of Public Health, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
- The Key Laboratory of Modern Toxicology of Ministry of Education, Department of Genetic Toxicology, School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Meilin Wang
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Department of Environmental Genomics, School of Public Health, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
- The Key Laboratory of Modern Toxicology of Ministry of Education, Department of Genetic Toxicology, School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, China
- Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, Suzhou, China
| | - Gong Cheng
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University & Jiangsu Province People's Hospital, 300 Guangzhou Road, Nanjing, 210029, China.
| | - Mulong Du
- Department of Biostatistics, School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, China.
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Harvard University, 655 Huntington Avenue, Boston, MA, 02115, USA.
| |
Collapse
|
19
|
Eng SE, Basasie B, Lam A, John Semmes O, Troyer DA, Clarke GD, Sunnapwar AG, Leach RJ, Johnson-Pais TL, Sokoll LJ, Chan DW, Tosoian JJ, Siddiqui J, Chinnaiyan AM, Thompson IM, Boutros PC, Liss MA. Prospective comparison of restriction spectrum imaging and non-invasive biomarkers to predict upgrading on active surveillance prostate biopsy. Prostate Cancer Prostatic Dis 2024; 27:65-72. [PMID: 36097168 DOI: 10.1038/s41391-022-00591-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/10/2022] [Accepted: 08/24/2022] [Indexed: 11/08/2022]
Abstract
BACKGROUND Protocol-based active surveillance (AS) biopsies have led to poor compliance. To move to risk-based protocols, more accurate imaging biomarkers are needed to predict upgrading on AS prostate biopsy. We compared restriction spectrum imaging (RSI-MRI) generated signal maps as a biomarker to other available non-invasive biomarkers to predict upgrading or reclassification on an AS biopsy. METHODS We prospectively enrolled men on prostate cancer AS undergoing repeat biopsy from January 2016 to June 2019 to obtain an MRI and biomarkers to predict upgrading. Subjects underwent a prostate multiparametric MRI and a short duration, diffusion-weighted enhanced MRI called RSI to generate a restricted signal map along with evaluation of 30 biomarkers (14 clinico-epidemiologic features, 9 molecular biomarkers, and 7 radiologic-associated features). Our primary outcome was upgrading or reclassification on subsequent AS prostate biopsy. Statistical analysis included operating characteristic improvement using AUROC and AUPRC. RESULTS The individual biomarker with the highest area under the receiver operator characteristic curve (AUC) was RSI-MRI (AUC = 0.84; 95% CI: 0.71-0.96). The best non-imaging biomarker was prostate volume-corrected Prostate Health Index density (PHI, AUC = 0.68; 95% CI: 0.53-0.82). Non-imaging biomarkers had a negligible effect on predicting upgrading at the next biopsy but did improve predictions of overall time to progression in AS. CONCLUSIONS RSI-MRI, PIRADS, and PHI could improve the predictive ability to detect upgrading in AS. The strongest predictor of clinically significant prostate cancer on AS biopsy was RSI-MRI signal output.
Collapse
Affiliation(s)
- Stefan E Eng
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA
- Institute for Precision Health, UCLA, Los Angeles, CA, USA
- Department of Urology, UCLA, Los Angeles, CA, USA
| | - Benjamin Basasie
- Department of Urology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Alfonso Lam
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA
- Institute for Precision Health, UCLA, Los Angeles, CA, USA
- Department of Urology, UCLA, Los Angeles, CA, USA
| | - O John Semmes
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Dean A Troyer
- Department of Pathology, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Geoffrey D Clarke
- Research Imaging Institute, University of Texas Health San Antonio, San Antonio, TX, USA
- Department of Radiology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Abhijit G Sunnapwar
- Department of Radiology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Robin J Leach
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX, USA
| | | | - Lori J Sokoll
- Department of Pathology, Division of Clinical Chemistry, Johns Hopkins University, Baltimore, MD, USA
| | - Daniel W Chan
- Department of Pathology, Division of Clinical Chemistry, Johns Hopkins University, Baltimore, MD, USA
| | | | - Javed Siddiqui
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | | | | | - Paul C Boutros
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA.
- Institute for Precision Health, UCLA, Los Angeles, CA, USA.
- Department of Urology, UCLA, Los Angeles, CA, USA.
- Department of Human Genetics, UCLA, Los Angeles, CA, USA.
- Broad Stem Cell Research Center, UCLA, Los Angeles, CA, USA.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
| | - Michael A Liss
- Department of Urology, University of Texas Health San Antonio, San Antonio, TX, USA.
- Research Imaging Institute, University of Texas Health San Antonio, San Antonio, TX, USA.
- College of Pharmacy, University of Texas Austin, Austin, TX, USA.
- Department of Urology, South Texas Veterans Healthcare System, San Antonio, TX, USA.
| |
Collapse
|
20
|
Amini AE, Salari K. Incorporating Genetic Risk Into Prostate Cancer Care: Implications for Early Detection and Precision Oncology. JCO Precis Oncol 2024; 8:e2300560. [PMID: 38412389 DOI: 10.1200/po.23.00560] [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: 10/11/2023] [Revised: 12/12/2023] [Accepted: 01/08/2024] [Indexed: 02/29/2024] Open
Abstract
The availability and cost of germline and somatic genetic testing have dramatically improved over the past two decades, enabling precision medicine approaches in oncology, with significant implications for prostate cancer (PCa) care. Roughly 12% of individuals with advanced disease are carriers of rare pathogenic germline variants that predispose to particularly aggressive and earlier-onset disease. Several of these variants are already established as clinically actionable by modern precision oncology therapeutics, while others may come to aid the selection of active surveillance, definitive local therapies, and systemic therapies. Concurrently, the number of common variants (ie, incorporated into polygenic risk scores) associated with PCa risk has continued to grow, but with several important considerations both at the intersection of race and ancestry and for early detection of aggressive disease. Family history has historically been used as a proxy for this inherited genetic risk of PCa, but recently emerging evidence examining this relation has shifted our understanding of how best to leverage this tool in PCa care. This review seeks to clarify and contextualize the existing and emerging precision oncology paradigms that use inherited genetic risk in PCa care, for both early detection and localized disease management.
Collapse
Affiliation(s)
- Andrew E Amini
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Keyan Salari
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
| |
Collapse
|
21
|
Kim SJ, Park M, Choi A, Yoo S. Microbiome and Prostate Cancer: Emerging Diagnostic and Therapeutic Opportunities. Pharmaceuticals (Basel) 2024; 17:112. [PMID: 38256945 PMCID: PMC10819128 DOI: 10.3390/ph17010112] [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: 12/11/2023] [Revised: 01/06/2024] [Accepted: 01/07/2024] [Indexed: 01/24/2024] Open
Abstract
This review systematically addresses the correlation between the microbiome and prostate cancer and explores its diagnostic and therapeutic implications. Recent research has indicated an association between the urinary and gut microbiome composition and prostate cancer incidence and progression. Specifically, the urinary microbiome is a potential non-invasive biomarker for early detection and risk evaluation, with altered microbial profiles in prostate cancer patients. This represents an advancement in non-invasive diagnostic approaches to prostate cancer. The role of the gut microbiome in the efficacy of various cancer therapies has recently gained attention. Gut microbiota variations can affect the metabolism and effectiveness of standard treatment modalities, including chemotherapy, immunotherapy, and hormone therapy. This review explores the potential of gut microbiome modification through dietary interventions, prebiotics, probiotics, and fecal microbiota transplantation for improving the treatment response and mitigating adverse effects. Moreover, this review discusses the potential of microbiome profiling for patient stratification and personalized treatment strategies. While the current research identifies the pivotal role of the microbiome in prostate cancer, it also highlights the necessity for further investigations to fully understand these complex interactions and their practical applications in improving patient outcomes in prostate cancer management.
Collapse
Affiliation(s)
- Sung Jin Kim
- Department of Urology, Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung 25440, Republic of Korea;
| | - Myungchan Park
- Department of Urology, Haeundae Paik Hospital, Inje University College of Medicine, Busan 47392, Republic of Korea;
| | - Ahnryul Choi
- Department of Biomedical Engineering, College of Medical Convergence, Catholic Kwandong University, Gangneung 25601, Republic of Korea
| | - Sangjun Yoo
- Department of Urology, SNU-SMG Boramae Medical Center, Seoul 07061, Republic of Korea
| |
Collapse
|
22
|
Xu L, Zhou G, Jiang W, Guan L, Zhao H. Leveraging genetic correlations and multiple populations to improve genetic risk prediction for non-European populations. RESEARCH SQUARE 2023:rs.3.rs-3741763. [PMID: 38234764 PMCID: PMC10793485 DOI: 10.21203/rs.3.rs-3741763/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
The disparity in genetic risk prediction accuracy between European and non-European individuals highlights a critical challenge in health inequality. To bridge this gap, we introduce JointPRS, a novel method that models multiple populations jointly to improve genetic risk predictions for non-European individuals. JointPRS has three key features. First, it encompasses all diverse populations to improve prediction accuracy, rather than relying solely on the target population with a singular auxiliary European group. Second, it autonomously estimates and leverages chromosome-wise cross-population genetic correlations to infer the effect sizes of genetic variants. Lastly, it provides an auto version that has comparable performance to the tuning version to accommodate the situation with no validation dataset. Through extensive simulations and real data applications to 22 quantitative traits and four binary traits in East Asian populations, nine quantitative traits and one binary trait in African populations, and four quantitative traits in South Asian populations, we demonstrate that JointPRS outperforms state-of-art methods, improving the prediction accuracy for both quantitative and binary traits in non-European populations.
Collapse
Affiliation(s)
- Leqi Xu
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - Geyu Zhou
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - Wei Jiang
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - Leying Guan
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Hongyu Zhao
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| |
Collapse
|
23
|
Khan A, Shang N, Nestor JG, Weng C, Hripcsak G, Harris PC, Gharavi AG, Kiryluk K. Polygenic risk alters the penetrance of monogenic kidney disease. Nat Commun 2023; 14:8318. [PMID: 38097619 PMCID: PMC10721887 DOI: 10.1038/s41467-023-43878-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 11/22/2023] [Indexed: 12/17/2023] Open
Abstract
Chronic kidney disease (CKD) is determined by an interplay of monogenic, polygenic, and environmental risks. Autosomal dominant polycystic kidney disease (ADPKD) and COL4A-associated nephropathy (COL4A-AN) represent the most common forms of monogenic kidney diseases. These disorders have incomplete penetrance and variable expressivity, and we hypothesize that polygenic factors explain some of this variability. By combining SNP array, exome/genome sequence, and electronic health record data from the UK Biobank and All-of-Us cohorts, we demonstrate that the genome-wide polygenic score (GPS) significantly predicts CKD among ADPKD monogenic variant carriers. Compared to the middle tertile of the GPS for noncarriers, ADPKD variant carriers in the top tertile have a 54-fold increased risk of CKD, while ADPKD variant carriers in the bottom tertile have only a 3-fold increased risk of CKD. Similarly, the GPS significantly predicts CKD in COL4A-AN carriers. The carriers in the top tertile of the GPS have a 2.5-fold higher risk of CKD, while the risk for carriers in the bottom tertile is not different from the average population risk. These results suggest that accounting for polygenic risk improves risk stratification in monogenic kidney disease.
Collapse
Affiliation(s)
- Atlas Khan
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Ning Shang
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Jordan G Nestor
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Chunhua Weng
- Department of Biomedical Informatics, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - George Hripcsak
- Department of Biomedical Informatics, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Peter C Harris
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Ali G Gharavi
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Krzysztof Kiryluk
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA.
| |
Collapse
|
24
|
Pan J, Sun J, Goncalves I, Kessler M, Hao Y, Engström G. Red cell distribution width and its polygenic score in relation to mortality and cardiometabolic outcomes. Front Cardiovasc Med 2023; 10:1294218. [PMID: 38054099 PMCID: PMC10694461 DOI: 10.3389/fcvm.2023.1294218] [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: 09/14/2023] [Accepted: 11/03/2023] [Indexed: 12/07/2023] Open
Abstract
Introduction Elevated red cell distribution width (RDW) has been associated with a range of health outcomes. This study aims to examine prognostic and etiological roles of RDW levels, both phenotypic and genetic predisposition, in predicting cardiovascular outcomes, diabetes, chronic kidney disease (CKD) and mortality. Methods We studied 27,141 middle-aged adults from the Malmö Diet and Cancer study (MDCS) with a mean follow up of 21 years. RDW was measured with a hematology analyzer on whole blood samples. Polygenic scores for RDW (PGS-RDW) were constructed for each participant using genetic data in MDCS and published summary statistics from genome-wide association study of RDW (n = 408,112). Cox proportional hazards regression was used to assess associations between RDW, PGS-RDW and cardiovascular outcomes, diabetes, CKD and mortality, respectively. Results PGS-RDW was significantly associated with RDW (Pearson's correlation coefficient = 0.133, p < 0.001). RDW was significantly associated with incidence of stroke (hazard ratio (HR) per 1 standard deviation = 1.06, 95% confidence interval (CI): 1.02-1.10, p = 0.003), atrial fibrillation (HR = 1.09, 95% CI: 1.06-1.12, p < 0.001), heart failure (HR = 1.13, 95% CI: 1.08-1.19, p < 0.001), venous thromboembolism (HR = 1.21, 95% CI: 1.15-1.28, p < 0.001), diabetes (HR = 0.87, 95% CI: 0.84-0.90, p < 0.001), CKD (HR = 1.08, 95% CI: 1.03-1.13, p = 0.004) and all-cause mortality (HR = 1.18, 95% CI: 1.16-1.20, p < 0.001). However, PGS-RDW was significantly associated with incidence of diabetes (HR = 0.96, 95% CI: 0.94-0.99, p = 0.01), but not with any other tested outcomes. Discussion RDW is associated with mortality and incidence of cardiovascular diseases, but a significant association between genetically determined RDW and incident cardiovascular diseases were not observed. However, both RDW and PGS-RDW were inversely associated with incidence of diabetes, suggesting a putative causal relationship. The relationship with incidence of diabetes needs to be further studied.
Collapse
Affiliation(s)
- Jingxue Pan
- Division of Child Healthcare, Department of Paediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Jiangming Sun
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | | | | | - Yan Hao
- Division of Child Healthcare, Department of Paediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gunnar Engström
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Regeneron Genetics Center, Tarrytown, NY, United States
| |
Collapse
|
25
|
Koch S, Schmidtke J, Krawczak M, Caliebe A. Clinical utility of polygenic risk scores: a critical 2023 appraisal. J Community Genet 2023; 14:471-487. [PMID: 37133683 PMCID: PMC10576695 DOI: 10.1007/s12687-023-00645-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 03/31/2023] [Indexed: 05/04/2023] Open
Abstract
Since their first appearance in the context of schizophrenia and bipolar disorder in 2009, polygenic risk scores (PRSs) have been described for a large number of common complex diseases. However, the clinical utility of PRSs in disease risk assessment or therapeutic decision making is likely limited because PRSs usually only account for the heritable component of a trait and ignore the etiological role of environment and lifestyle. We surveyed the current state of PRSs for various diseases, including breast cancer, diabetes, prostate cancer, coronary artery disease, and Parkinson disease, with an extra focus upon the potential improvement of clinical scores by their combination with PRSs. We observed that the diagnostic and prognostic performance of PRSs alone is consistently low, as expected. Moreover, combining a PRS with a clinical score at best led to moderate improvement of the power of either risk marker. Despite the large number of PRSs reported in the scientific literature, prospective studies of their clinical utility, particularly of the PRS-associated improvement of standard screening or therapeutic procedures, are still rare. In conclusion, the benefit to individual patients or the health care system in general of PRS-based extensions of existing diagnostic or treatment regimens is still difficult to judge.
Collapse
Affiliation(s)
- Sebastian Koch
- Institut für Medizinische Informatik und Statistik, Christian-Albrechts-Universität zu Kiel, Universitätsklinikum Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Jörg Schmidtke
- Amedes MVZ Wagnerstibbe, Hannover, Germany
- Institut für Humangenetik, Medizinische Hochschule Hannover, Hannover, Germany
| | - Michael Krawczak
- Institut für Medizinische Informatik und Statistik, Christian-Albrechts-Universität zu Kiel, Universitätsklinikum Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Amke Caliebe
- Institut für Medizinische Informatik und Statistik, Christian-Albrechts-Universität zu Kiel, Universitätsklinikum Schleswig-Holstein Campus Kiel, Kiel, Germany.
| |
Collapse
|
26
|
Le T, Rojas PS, Fakunle M, Huang FW. Racial disparity in the genomics of precision oncology of prostate cancer. Cancer Rep (Hoboken) 2023; 6 Suppl 1:e1867. [PMID: 37565547 PMCID: PMC10440844 DOI: 10.1002/cnr2.1867] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 06/15/2023] [Accepted: 06/30/2023] [Indexed: 08/12/2023] Open
Abstract
BACKGROUND Significant racial disparities in prostate cancer incidence and mortality have been reported between African American Men (AAM), who are at increased risk for prostate cancer, and European American Men (EAM). In most of the studies carried out on prostate cancer, this population is underrepresented. With the advancement of genome-wide association studies, several genetic predictor models of prostate cancer risk have been elaborated, as well as numerous studies that identify both germline and somatic mutations with clinical utility. RECENT FINDINGS Despite significant advances, the AAM population continues to be underrepresented in genomic studies, which can limit generalizability and potentially widen disparities. Here we outline racial disparities in currently available genomic applications that are used to estimate the risk of individuals developing prostate cancer and to identify personalized oncology treatment strategies. While the incidence and mortality of prostate cancer are different between AAM and EAM, samples from AAM remain to be unrepresented in different studies. CONCLUSION This disparity impacts the available genomic data on prostate cancer. As a result, the disparity can limit the predictive utility of the genomic applications and may lead to the widening of the existing disparities. More studies with substantially higher recruitment and engagement of African American patients are necessary to overcome this disparity.
Collapse
Affiliation(s)
- Tu Le
- Division of Hematology and Oncology, Department of MedicineUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Division of Hematology and Oncology, Department of MedicineSan Francisco Veterans Affairs Medical CenterSan FranciscoCaliforniaUSA
| | - Pilar Soto Rojas
- Division of Hematology and Oncology, Department of MedicineUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of OncologyHospital Universitario Virgen MacarenaSevilleSpain
| | - Mary Fakunle
- Department of UrologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Franklin W. Huang
- Division of Hematology and Oncology, Department of MedicineUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Division of Hematology and Oncology, Department of MedicineSan Francisco Veterans Affairs Medical CenterSan FranciscoCaliforniaUSA
- Department of UrologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Chan Zuckerberg BiohubSan FranciscoCaliforniaUSA
- Institute for Human GeneticsUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Bakar Computational Health Sciences InstituteUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Benioff Initiative for Prostate Cancer ResearchUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| |
Collapse
|
27
|
Sekimitsu S, Xiang D, Smith SL, Curran K, Elze T, Friedman DS, Foster PJ, Luo Y, Pasquale LR, Peto T, Segrè AV, Shweikh Y, Warwick A, Zhao Y, Wiggs JL, Zebardast N. Deep Ocular Phenotyping Across Primary Open-Angle Glaucoma Genetic Burden. JAMA Ophthalmol 2023; 141:891-899. [PMID: 37589995 PMCID: PMC10436188 DOI: 10.1001/jamaophthalmol.2023.3645] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/25/2023] [Indexed: 08/18/2023]
Abstract
Importance Better understanding of primary open-angle glaucoma (POAG) genetics could enable timely screening and promote individualized disease risk prognostication. Objective To evaluate phenotypic features across genetic burden for POAG. Design, Setting, and Participants This was a cross-sectional, population-based study conducted from 2006 to 2010. Included participants were individuals from the UK Biobank aged 40 to 69 years. Individuals with non-POAG forms of glaucoma were excluded from the analysis. Data were statistically analyzed from October 2022 to January 2023. Main Outcomes and Measures POAG prevalence based on structural coding, self-reports, and glaucoma-related traits. Results Among 407 667 participants (mean [SD] age, 56.3 [8.1] years; 219 183 majority sex [53.8%]) were 14 171 POAG cases. Area under receiver operating characteristic curve for POAG detection was 0.748 in a model including polygenic risk score (PRS), age, sex, and ancestry. POAG prevalence in the highest decile of PRS was 7.4% (3005 of 40 644) vs 1.3% (544 of 40 795) in lowest decile (P < .001). A 1-SD increase in PRS was associated with 1.74 times higher odds of POAG (95% CI, 1.71-1.77), a 0.61-mm Hg increase in corneal-compensated intraocular pressure (IOP; 95% CI, 0.59-0.64), a -0.09-mm Hg decrease in corneal hysteresis (95% CI, -0.10 to -0.08), a 0.08-mm Hg increase in corneal resistance factor (95% CI, 0.06-0.09), and a -0.08-diopter decrease in spherical equivalent (95% CI, -0.11 to -0.07; P < .001 for all). A 1-SD increase in PRS was associated with a thinning of the macula-region retinal nerve fiber layer (mRNFL) of 0.14 μm and macular ganglion cell complex (GCC) of 0.26 μm (P < .001 for both). In the subset of individuals with fundus photographs, a 1-SD increase in PRS was associated with 1.42 times higher odds of suspicious optic disc features (95% CI, 1.19-1.69) and a 0.013 increase in cup-disc ratio (CDR; 95% CI, 0.012-0.014; P < .001 for both). A total of 22 of 5193 fundus photographs (0.4%) in decile 10 had disc hemorrhages, and 27 of 5257 (0.5%) had suspicious optic disc features compared with 9 of 5158 (0.2%) and 10 of 5219 (0.2%), respectively, in decile 1 (P < .001 for both). CDR in decile 10 was 0.46 compared with 0.41 in decile 1 (P < .001). Conclusion and Relevance Results suggest that PRS identified a group of individuals at substantially higher risk for POAG. Higher genetic risk was associated with more advanced disease, namely higher CDR and corneal-compensated IOP, thinner mRNFL, and thinner GCC. Associations with POAG PRS and corneal hysteresis and greater prevalence of disc hemorrhages were identified. These results suggest that genetic risk is an increasingly important parameter for risk stratification to consider in clinical practice.
Collapse
Affiliation(s)
| | - David Xiang
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston
- Harvard Medical School, Boston, Massachusetts
| | | | - Katie Curran
- Centre for Public Health, Queen’s University Belfast, Belfast, United Kingdom
| | - Tobias Elze
- Schepens Eye Research Institute, Harvard Medical School, Boston, Massachusetts
| | - David S. Friedman
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston
| | - Paul J. Foster
- NIHR Biomedical Research Centre, Moorfields Eye Hospital & UCL Institute of Ophthalmology, London, United Kingdom
| | - Yuyang Luo
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston
- Ocular Genomics Institute, Harvard Medical School, Boston, Massachusetts
| | - Louis R. Pasquale
- Icahn School of Medicine at Mount Sinai, Department of Ophthalmology, New York, New York
| | - Tunde Peto
- Centre for Public Health, Queen’s University Belfast, Belfast, United Kingdom
| | - Ayellet V. Segrè
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston
- Ocular Genomics Institute, Harvard Medical School, Boston, Massachusetts
| | - Yusrah Shweikh
- Sussex Eye Hospital, University Hospitals Sussex NHS Foundation Trust, Sussex, United Kingdom
| | - Alasdair Warwick
- University College London, Institute of Cardiovascular Science, London, United Kingdom
- Medical Retina Service, Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom
| | - Yan Zhao
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston
- Ocular Genomics Institute, Harvard Medical School, Boston, Massachusetts
| | - Janey L. Wiggs
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston
- Ocular Genomics Institute, Harvard Medical School, Boston, Massachusetts
| | - Nazlee Zebardast
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston
| |
Collapse
|
28
|
Akdeniz BC, Mattingsdal M, Dominguez-Valentin M, Frei O, Shadrin A, Puustusmaa M, Saar R, Sõber S, Møller P, Andreassen OA, Padrik P, Hovig E. A Breast Cancer Polygenic Risk Score Is Feasible for Risk Stratification in the Norwegian Population. Cancers (Basel) 2023; 15:4124. [PMID: 37627152 PMCID: PMC10452897 DOI: 10.3390/cancers15164124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/11/2023] [Accepted: 08/13/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Statistical associations of numerous single nucleotide polymorphisms with breast cancer (BC) have been identified in genome-wide association studies (GWAS). Recent evidence suggests that a Polygenic Risk Score (PRS) can be a useful risk stratification instrument for a BC screening strategy, and a PRS test has been developed for clinical use. The performance of the PRS is yet unknown in the Norwegian population. AIM To evaluate the performance of PRS models for BC in a Norwegian dataset. METHODS We investigated a sample of 1053 BC cases and 7094 controls from different regions of Norway. PRS values were calculated using four PRS models, and their performance was evaluated by the area under the curve (AUC) and the odds ratio (OR). The effect of the PRS on the age of onset of BC was determined by a Cox regression model, and the lifetime absolute risk of developing BC was calculated using the iCare tool. RESULTS The best performing PRS model included 3820 SNPs, which yielded an AUC = 0.625 and an OR = 1.567 per one standard deviation increase. The PRS values of the samples correlate with an increased risk of BC, with a hazard ratio of 1.494 per one standard deviation increase (95% confidence interval of 1.406-1.588). The individuals in the highest decile of the PRS have at least twice the risk of developing BC compared to the individuals with a median PRS. The results in this study with Norwegian samples are coherent with the findings in the study conducted using Estonian and UK Biobank samples. CONCLUSION The previously validated PRS models have a similar observed accuracy in the Norwegian data as in the UK and Estonian populations. A PRS provides a meaningful association with the age of onset of BC and lifetime risk. Therefore, as suggested in Estonia, a PRS may also be integrated into the screening strategy for BC in Norway.
Collapse
Affiliation(s)
- Bayram Cevdet Akdeniz
- Center for Bioinformatics, Department of Informatics, University of Oslo, 0313 Oslo, Norway (E.H.)
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, University of Oslo, 4956 Oslo, Norway (O.A.A.)
| | - Morten Mattingsdal
- Center for Bioinformatics, Department of Informatics, University of Oslo, 0313 Oslo, Norway (E.H.)
- Department of Medical Research, Vestre Viken Hospital Trust, Bærum Hospital, 1346 Gjettum, Norway
| | - Mev Dominguez-Valentin
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, 0424 Oslo, Norway (P.M.)
| | - Oleksandr Frei
- Center for Bioinformatics, Department of Informatics, University of Oslo, 0313 Oslo, Norway (E.H.)
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, University of Oslo, 4956 Oslo, Norway (O.A.A.)
| | - Alexey Shadrin
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, University of Oslo, 4956 Oslo, Norway (O.A.A.)
| | | | - Regina Saar
- OÜ Antegenes, 50603 Tartu, Estonia; (M.P.); (R.S.)
| | - Siim Sõber
- OÜ Antegenes, 50603 Tartu, Estonia; (M.P.); (R.S.)
| | - Pål Møller
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, 0424 Oslo, Norway (P.M.)
| | - Ole A. Andreassen
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, University of Oslo, 4956 Oslo, Norway (O.A.A.)
| | | | - Eivind Hovig
- Center for Bioinformatics, Department of Informatics, University of Oslo, 0313 Oslo, Norway (E.H.)
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, 0424 Oslo, Norway (P.M.)
| |
Collapse
|
29
|
Lui AJ, Pagadala MS, Zhong AY, Lynch J, Karunamuni R, Lee KM, Plym A, Rose BS, Carter H, Kibel AS, DuVall SL, Gaziano JM, Panizzon MS, Hauger RL, Seibert TM. Agent Orange exposure and prostate cancer risk in the Million Veteran Program. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.14.23291413. [PMID: 37398205 PMCID: PMC10312838 DOI: 10.1101/2023.06.14.23291413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Purpose Exposure to Agent Orange, a known carcinogen, might increase risk of prostate cancer (PCa). We sought to investigate the association of Agent Orange exposure and PCa risk when accounting for race/ethnicity, family history, and genetic risk in a diverse population of US Vietnam War veterans. Methods & Materials This study utilized the Million Veteran Program (MVP), a national, population-based cohort study of United States military veterans conducted 2011-2021 with 590,750 male participants available for analysis. Agent Orange exposure was obtained using records from the Department of Veterans Affairs (VA) using the US government definition of Agent Orange exposure: active service in Vietnam while Agent Orange was in use. Only veterans who were on active duty (anywhere in the world) during the Vietnam War were included in this analysis (211,180 participants). Genetic risk was assessed via a previously validated polygenic hazard score calculated from genotype data. Age at diagnosis of any PCa, diagnosis of metastatic PCa, and death from PCa were assessed via Cox proportional hazards models. Results Exposure to Agent Orange was associated with increased PCa diagnosis (HR 1.04, 95% CI 1.01-1.06, p=0.003), primarily among Non-Hispanic White men (HR 1.09, 95% CI 1.06- 1.12, p<0.001). When accounting for race/ethnicity and family history, Agent Orange exposure remained an independent risk factor for PCa diagnosis (HR 1.06, 95% CI 1.04-1.09, p<0.05). Univariable associations of Agent Orange exposure with PCa metastasis (HR 1.08, 95% CI 0.99-1.17) and PCa death (HR 1.02, 95% CI 0.84-1.22) did not reach significance on multivariable analysis. Similar results were found when accounting for polygenic hazard score. Conclusions Among US Vietnam War veterans, Agent Orange exposure is an independent risk factor for PCa diagnosis, though associations with PCa metastasis or death are unclear when accounting for race/ethnicity, family history, and/or polygenic risk.
Collapse
|
30
|
Shortall J, Vasquez Osorio E, Green A, McWilliam A, Elumalai T, Reeves K, Johnson-Hart C, Beasley W, Hoskin P, Choudhury A, van Herk M. Dose outside of the prostate is associated with improved outcomes for high-risk prostate cancer patients treated with brachytherapy boost. Front Oncol 2023; 13:1200676. [PMID: 37397380 PMCID: PMC10311256 DOI: 10.3389/fonc.2023.1200676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 05/31/2023] [Indexed: 07/04/2023] Open
Abstract
Background One in three high-risk prostate cancer patients treated with radiotherapy recur. Detection of lymph node metastasis and microscopic disease spread using conventional imaging is poor, and many patients are under-treated due to suboptimal seminal vesicle or lymph node irradiation. We use Image Based Data Mining (IBDM) to investigate association between dose distributions, and prognostic variables and biochemical recurrence (BCR) in prostate cancer patients treated with radiotherapy. We further test whether including dose information in risk-stratification models improves performance. Method Planning CTs, dose distributions and clinical information were collected for 612 high-risk prostate cancer patients treated with conformal hypo-fractionated radiotherapy, intensity modulated radiotherapy (IMRT), or IMRT plus a single fraction high dose rate (HDR) brachytherapy boost. Dose distributions (including HDR boost) of all studied patients were mapped to a reference anatomy using the prostate delineations. Regions where dose distributions significantly differed between patients that did and did-not experience BCR were assessed voxel-wise using 1) a binary endpoint of BCR at four-years (dose only) and 2) Cox-IBDM (dose and prognostic variables). Regions where dose was associated with outcome were identified. Cox proportional-hazard models with and without region dose information were produced and the Akaike Information Criterion (AIC) was used to assess model performance. Results No significant regions were observed for patients treated with hypo-fractionated radiotherapy or IMRT. Regions outside the target where higher dose was associated with lower BCR were observed for patients treated with brachytherapy boost. Cox-IBDM revealed that dose response was influenced by age and T-stage. A region at the seminal vesicle tips was identified in binary- and Cox-IBDM. Including the mean dose in this region in a risk-stratification model (hazard ratio=0.84, p=0.005) significantly reduced AIC values (p=0.019), indicating superior performance, compared with prognostic variables only. The region dose was lower in the brachytherapy boost patients compared with the external beam cohorts supporting the occurrence of marginal misses. Conclusion Association was identified between BCR and dose outside of the target region in high-risk prostate cancer patients treated with IMRT plus brachytherapy boost. We show, for the first-time, that the importance of irradiating this region is linked to prognostic variables.
Collapse
Affiliation(s)
- Jane Shortall
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Eliana Vasquez Osorio
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Andrew Green
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Alan McWilliam
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
- Department of Radiotherapy Related Research, The Christie National Health Service (NHS) Foundation Trust, Manchester, United Kingdom
| | - Thriaviyam Elumalai
- Department of Radiotherapy Related Research, The Christie National Health Service (NHS) Foundation Trust, Manchester, United Kingdom
| | - Kimberley Reeves
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Corinne Johnson-Hart
- Department of Radiotherapy Related Research, The Christie National Health Service (NHS) Foundation Trust, Manchester, United Kingdom
| | - William Beasley
- Department of Radiotherapy Related Research, The Christie National Health Service (NHS) Foundation Trust, Manchester, United Kingdom
| | - Peter Hoskin
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
- Department of Radiotherapy Related Research, The Christie National Health Service (NHS) Foundation Trust, Manchester, United Kingdom
| | - Ananya Choudhury
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
- Department of Radiotherapy Related Research, The Christie National Health Service (NHS) Foundation Trust, Manchester, United Kingdom
| | - Marcel van Herk
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
31
|
Phulka JS, Ashraf M, Bajwa BK, Pare G, Laksman Z. Current State and Future of Polygenic Risk Scores in Cardiometabolic Disease: A Scoping Review. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2023; 16:286-313. [PMID: 37035923 DOI: 10.1161/circgen.122.003834] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
A polygenic risk score (PRS) is derived from a genome-wide association study and represents an aggregate of thousands of single-nucleotide polymorphisms that provide a baseline estimate of an individual's genetic risk for a specific disease or trait at birth. However, it remains unclear how PRSs can be used in clinical practice. We provide an overview of the PRSs related to cardiometabolic disease and discuss the evidence supporting their clinical applications and limitations. The Preferred Reporting Items For Systematic Reviews and Meta-Analysis Extension for Scoping Reviews protocol was used to conduct a scoping review of the MEDLINE, EMBASE, and CENTRAL databases. Across the 4863 studies screened, 82 articles met the inclusion criteria. The most common PRS related to coronary artery disease, followed by hypertension and cerebrovascular disease. Limited ancestral diversity was observed in the study sample populations. Most studies included only individuals of European ancestry. The predictive performance of most PRSs was similar to or superior to traditional risk factors. More than half of the included studies reported an integrated risk model combining a derived PRS and clinical risk tools such as the Framingham Risk Score and Pooled Cohort Equations. The inclusion of a PRS into a clinical risk model tended to improve predictive accuracy consistently. This scoping review is the first of its kind and reports strong evidence for the clinical utility of PRSs in coronary artery disease, hypertension, cerebrovascular disease, and atrial fibrillation. However, most PRSs are generated in cohorts of European ancestry, which likely contributes to a lack of PRS transferability across different ancestral groups. Future prospective studies should focus on further establishing the clinical utility of PRSs and ensuring diversity is incorporated into genome-wide association study cohorts.
Collapse
Affiliation(s)
- Jobanjit S Phulka
- Heart Rhythm Services & Center for Cardiovascular Innovation, Division of Cardiology, University of British Columbia, Vancouver (J.S.P., M.A., B.K.B., Z.L.)
| | - Mishal Ashraf
- Heart Rhythm Services & Center for Cardiovascular Innovation, Division of Cardiology, University of British Columbia, Vancouver (J.S.P., M.A., B.K.B., Z.L.)
| | - Beenu K Bajwa
- Heart Rhythm Services & Center for Cardiovascular Innovation, Division of Cardiology, University of British Columbia, Vancouver (J.S.P., M.A., B.K.B., Z.L.)
| | - Guillaume Pare
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute; Thrombosis and Atherosclerosis Research Institute, Department of Health Research Methods, Evidence, and Impact, Department of Pathology & Molecular Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON, Canada (G.P.)
| | - Zachary Laksman
- Heart Rhythm Services & Center for Cardiovascular Innovation, Division of Cardiology, University of British Columbia, Vancouver (J.S.P., M.A., B.K.B., Z.L.)
| |
Collapse
|
32
|
Schirle L, Samuels DC, Faucon A, Cox NJ, Bruehl S. Polygenic Contributions to Chronic Overlapping Pain Conditions in a Large Electronic Health Record Sample. THE JOURNAL OF PAIN 2023; 24:1056-1068. [PMID: 36736868 PMCID: PMC10257768 DOI: 10.1016/j.jpain.2023.01.018] [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: 04/02/2022] [Revised: 01/11/2023] [Accepted: 01/23/2023] [Indexed: 02/05/2023]
Abstract
Chronic overlapping pain conditions (COPCs) are believed to share common etiological mechanisms involving central sensitization. Genetic and environmental factors putatively combine to influence susceptibility to central sensitization and COPCs. This study employed a genome-wide polygenic risk score approach to evaluate genetic influences on 8 common COPCs. COPCs were identified by International Classification of Disease codes in Vanderbilt's deidentified clinical biorepository (BioVU), with each COPC condition empirically weighted for the level of central sensitization based on prior work. A centralized pain score (CPS) was calculated for 55,340 individuals by summing the weighted number of COPCs. Overall, 12,502 individuals (22.6%) were diagnosed with at least 1 COPC, with females exhibiting nearly twice the mean CPS as males. To assess the genetic influence on centralized pain in COPCs, 6 pain polygenic risk scores (PRSs) were developed using UK Biobank data to predict 6 pain criteria (no pain, neck/shoulder, abdomen, hip, knee, low back pain). These PRSs were then deployed in the BioVU cohort to test for association with CPS. In regression models adjusted for age, sex, and BMI, all pain PRSs except hip pain were significantly associated with CPS. Our findings support a shared polygenic influence across COPCs potentially involving central sensitization mechanisms. PERSPECTIVE: This study used a polygenic risk score approach to investigate genetic influences on chronic overlapping pain conditions. Significant findings in this study provide evidence supporting previous hypotheses that a shared polygenic influence involving central sensitization may underly chronic overlapping pain conditions and can guide future biomarker and risk assessment research.
Collapse
Affiliation(s)
- Lori Schirle
- Vanderbilt University School of Nursing, Nashville, Tennessee.
| | - David C Samuels
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee; Vanderbilt Genetics Institute, Nashville, Tennessee
| | | | - Nancy J Cox
- Vanderbilt Genetics Institute, Nashville, Tennessee; Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Stephen Bruehl
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
33
|
Plym A, Zhang Y, Stopsack KH, Delcoigne B, Wiklund F, Haiman C, Kenfield SA, Kibel AS, Giovannucci E, Penney KL, Mucci LA. A Healthy Lifestyle in Men at Increased Genetic Risk for Prostate Cancer. Eur Urol 2023; 83:343-351. [PMID: 35637041 DOI: 10.1016/j.eururo.2022.05.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/20/2022] [Accepted: 05/10/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Prostate cancer is the most heritable cancer. There is a need to identify possible modifiable factors for men at an increased risk of prostate cancer due to genetic factors. OBJECTIVE To examine whether men at an increased genetic risk of prostate cancer can offset their risk of disease or disease progression by adhering to a healthy lifestyle. DESIGN, SETTING, AND PARTICIPANTS We prospectively followed 12 411 genotyped men in the Health Professionals Follow-up Study (1993-2019) and the Physicians' Health Study (1983-2010). Genetic risk of prostate cancer was quantified using a polygenic risk score (PRS). A healthy lifestyle was defined by healthy weight, vigorous physical activity, not smoking, and a healthy diet. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Overall and lethal prostate cancer events (metastatic disease/prostate cancer-specific death) were analyzed using time-to-event analyses estimating hazard ratios (HRs) and lifetime risks. RESULTS AND LIMITATIONS During 27 yr of follow-up, 3005 overall prostate cancer and 435 lethal prostate cancer events were observed. The PRS enabled risk stratification not only for overall prostate cancer, but also for lethal disease with a four-fold difference between men in the highest and lowest quartiles (HR, 4.32; 95% confidence interval [CI], 3.16-5.89). Among men in the highest PRS quartile, adhering to a healthy lifestyle was associated with a decreased rate of lethal prostate cancer (HR, 0.55; 95% CI, 0.36-0.86) compared with having an unhealthy lifestyle, translating to a lifetime risk of 1.6% (95% CI, 0.8-3.1%) among the healthy and 5.3% (95% CI, 3.6-7.8%) among the unhealthy. Adhering to a healthy lifestyle was not associated with a decreased risk of overall prostate cancer. CONCLUSIONS Our findings suggest that a genetic predisposition for prostate cancer is not deterministic for a poor cancer outcome. Maintaining a healthy lifestyle may provide a way to offset the genetic risk of lethal prostate cancer. PATIENT SUMMARY This study examined whether the genetic risk of prostate cancer can be attenuated by a healthy lifestyle including a healthy weight, regular exercise, not smoking, and a healthy diet. We observed that adherence to a healthy lifestyle reduced the risk of metastatic disease and prostate cancer death among men at the highest genetic risk. We conclude that men at a high genetic risk of prostate cancer may benefit from adhering to a healthy lifestyle.
Collapse
Affiliation(s)
- Anna Plym
- Urology Division, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA; Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.
| | - Yiwen Zhang
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Konrad H Stopsack
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Bénédicte Delcoigne
- Division of Clinical Epidemiology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik Wiklund
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Christopher Haiman
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Stacey A Kenfield
- Departments of Urology and Epidemiology & Biostatistics, University of California, San Francisco, CA, USA
| | - Adam S Kibel
- Urology Division, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Edward Giovannucci
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Kathryn L Penney
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Lorelei A Mucci
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
34
|
Wang XY, Wang LL, Xu L, Liang SZ, Yu MC, Zhang QY, Dong QJ. Evaluation of polygenic risk score for risk prediction of gastric cancer. World J Gastrointest Oncol 2023; 15:276-285. [PMID: 36908320 PMCID: PMC9994049 DOI: 10.4251/wjgo.v15.i2.276] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/11/2023] [Accepted: 02/02/2023] [Indexed: 02/14/2023] Open
Abstract
Genetic variations are associated with individual susceptibility to gastric cancer. Recently, polygenic risk score (PRS) models have been established based on genetic variants to predict the risk of gastric cancer. To assess the accuracy of current PRS models in the risk prediction, a systematic review was conducted. A total of eight eligible studies consisted of 544842 participants were included for evaluation of the performance of PRS models. The overall accuracy was moderate with Area under the curve values ranging from 0.5600 to 0.7823. Incorporation of epidemiological factors or Helicobacter pylori (H. pylori) status increased the accuracy for risk prediction, while selection of single nucleotide polymorphism (SNP) and number of SNPs appeared to have little impact on the model performance. To further improve the accuracy of PRS models for risk prediction of gastric cancer, we summarized the association between gastric cancer risk and H. pylori genomic variations, cancer associated bacteria members in the gastric microbiome, discussed the potentials for performance improvement of PRS models with these microbial factors. Future studies on comprehensive PRS models established with human SNPs, epidemiological factors and microbial factors are indicated.
Collapse
Affiliation(s)
- Xiao-Yu Wang
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Li-Li Wang
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Lin Xu
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Shu-Zhen Liang
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Meng-Chao Yu
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Qiu-Yue Zhang
- Department of Clinical Laboratory, the Eighth Medical Center of the General Hospital of the People’s Liberation Army, Beijing 100000, China
| | - Quan-Jiang Dong
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong Province, China
| |
Collapse
|
35
|
Pagadala MS, Lynch J, Karunamuni R, Alba PR, Lee KM, Agiri FY, Anglin T, Carter H, Gaziano JM, Jasuja GK, Deka R, Rose BS, Panizzon MS, Hauger RL, Seibert TM. Polygenic risk of any, metastatic, and fatal prostate cancer in the Million Veteran Program. J Natl Cancer Inst 2023; 115:190-199. [PMID: 36305680 PMCID: PMC9905969 DOI: 10.1093/jnci/djac199] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 07/13/2022] [Accepted: 10/26/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Genetic scores may provide an objective measure of prostate cancer risk and thus inform screening decisions. We evaluated whether a polygenic hazard score based on 290 genetic variants (PHS290) is associated with prostate cancer risk in a diverse population, including Black men, who have higher average risk of prostate cancer death but are often treated as a homogeneously high-risk group. METHODS This was a retrospective analysis of the Million Veteran Program, a national, population-based cohort study of US military veterans conducted 2011-2021. Cox proportional hazards analyses tested for association of genetic and other risk factors (including self-reported race and ethnicity and family history) with age at death from prostate cancer, age at diagnosis of metastatic (nodal or distant) prostate cancer, and age at diagnosis of any prostate cancer. RESULTS A total of 590 750 male participants were included. Median age at last follow-up was 69 years. PHS290 was associated with fatal prostate cancer in the full cohort and for each racial and ethnic group (P < .001). Comparing men in the highest 20% of PHS290 with those in the lowest 20% (based on percentiles from an independent training cohort), the hazard ratio for fatal prostate cancer was 4.42 (95% confidence interval = 3.91 to 5.02). When accounting for guideline-recommended risk factors (family history, race, and ethnicity), PHS290 remained a strong independent predictor of any, metastatic, and fatal prostate cancer. CONCLUSIONS PHS290 stratified US veterans of diverse ancestry for lifetime risk of prostate cancer, including metastatic and fatal cancer. Predicting genetic risk of lethal prostate cancer with PHS290 might inform individualized decisions about prostate cancer screening.
Collapse
Affiliation(s)
- Meghana S Pagadala
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
- Medical Scientist Training Program, University of California San Diego, La Jolla, CA, USA
- Biomedical Science Program, University of California San Diego, La Jolla, CA, USA
| | - Julie Lynch
- VA Informatics and Computing Infrastructure, VA Salt Lake City Healthcare System (VINCI), Salt Lake City, UT, USA
- Department of Internal Medicine, Division of Epidemiology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Roshan Karunamuni
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, CA, USA
| | - Patrick R Alba
- VA Informatics and Computing Infrastructure, VA Salt Lake City Healthcare System (VINCI), Salt Lake City, UT, USA
- Department of Internal Medicine, Division of Epidemiology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Kyung Min Lee
- VA Informatics and Computing Infrastructure, VA Salt Lake City Healthcare System (VINCI), Salt Lake City, UT, USA
- Department of Internal Medicine, Division of Epidemiology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Fatai Y Agiri
- VA Informatics and Computing Infrastructure, VA Salt Lake City Healthcare System (VINCI), Salt Lake City, UT, USA
| | - Tori Anglin
- VA Informatics and Computing Infrastructure, VA Salt Lake City Healthcare System (VINCI), Salt Lake City, UT, USA
- Department of Internal Medicine, Division of Epidemiology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Hannah Carter
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - J Michael Gaziano
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Guneet Kaur Jasuja
- Center for Healthcare Organization and Implementation Research (CHOIR), VA Bedford Healthcare System, Bedford, MA, USA
- Section of General Internal Medicine, Boston University School of Medicine, Boston, MA, USA; Department of Urology, University of California San Diego, La Jolla, CA, USA
| | - Rishi Deka
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, CA, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Brent S Rose
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, CA, USA
- Section of General Internal Medicine, Boston University School of Medicine, Boston, MA, USA; Department of Urology, University of California San Diego, La Jolla, CA, USA
| | - Matthew S Panizzon
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
- Center for Behavioral Genetics of Aging, University of California San Diego, La Jolla, CA, USA
| | - Richard L Hauger
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
- Center for Behavioral Genetics of Aging, University of California San Diego, La Jolla, CA, USA
- Center of Excellence for Stress and Mental Health (CESAMH), VA San Diego Healthcare System, San Diego, CA, USA
| | - Tyler M Seibert
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, CA, USA
- Department of Radiology, University of California San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
36
|
The Combined Effect of Polygenic Risk Score and Prostate Health Index in Chinese Men Undergoing Prostate Biopsy. J Clin Med 2023; 12:jcm12041343. [PMID: 36835879 PMCID: PMC9960699 DOI: 10.3390/jcm12041343] [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: 09/30/2022] [Revised: 01/02/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Abstract
To date, the combined effect of polygenic risk score (PRS) and prostate health index (phi) on PCa diagnosis in men undergoing prostate biopsy has never been investigated. A total of 3166 patients who underwent initial prostate biopsy in three tertiary medical centers from August 2013 to March 2019 were included. PRS was calculated on the basis of the genotype of 102 reported East-Asian-specific risk variants. It was then evaluated in the univariable or multivariable logistic regression models that were internally validated using repeated 10-fold cross-validation. Discriminative performance was assessed by area under the receiver operating curve (AUC) and net reclassification improvement (NRI) index. Compared with men in the first quintile of age and family history adjusted PRS, those in the second, third, fourth, and fifth quintiles were 1.86 (odds ratio, 95% confidence interval (CI): 1.34-2.56), 2.07 (95%CI: 1.50-2.84), 3.26 (95%CI: 2.36-4.48), and 5.06 (95%CI: 3.68-6.97) times as likely to develop PCa (all p < 0.001). Adjustment for other clinical parameters yielded similar results. Among patients with prostate-specific antigen (PSA) at 2-10 ng/mL or 2-20 ng/mL, PRS still had an observable ability to differentiate PCa in the group of prostate health index (phi) at 27-36 (Ptrend < 0.05) or >36 (Ptrend ≤ 0.001). Notably, men with moderate phi (27-36) but highest PRS (top 20% percentile) would have a comparable risk of PCa (positive rate: 26.7% or 31.3%) than men with high phi (>36) but lowest PRS (bottom 20% percentile positive rate: 27.4% or 34.2%). The combined model of PRS, phi, and other clinical risk factors provided significantly better performance (AUC: 0.904, 95%CI: 0.887-0.921) than models without PRS. Adding PRS to clinical risk models could provide significant net benefit (NRI, from 8.6% to 27.6%), especially in those early onset patients (NRI, from 29.2% to 44.9%). PRS may provide additional predictive value over phi for PCa. The combination of PRS and phi that effectively captured both clinical and genetic PCa risk is clinically practical, even in patients with gray-zone PSA.
Collapse
|
37
|
Polygenic risk score for tumor aggressiveness and early-onset prostate cancer in Asians. Sci Rep 2023; 13:798. [PMID: 36646726 PMCID: PMC9842611 DOI: 10.1038/s41598-022-17515-2] [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: 05/26/2022] [Accepted: 07/26/2022] [Indexed: 01/18/2023] Open
Abstract
We attempted to assess the performance of an ethnic-specific polygenic risk score (PRS) designed from a Korean population to predict aggressive prostate cancer (PCa) and early-onset (age < 60). A PRS score comprised of 22 SNPs was computed in 3695 patients gathered from one of 4 tertiary centers in Korea. Males with biopsy or radical prostatectomy-proven PCa were included for analysis, collecting additional clinical parameters such as age, BMI, PSA, Gleason Group (GG), and staging. Patients were divided into 4 groups of PRS quartiles. Intergroup differences were assessed, as well as risk ratio and predictive performance based on GG using logistic regression analysis and AUC. No significant intergroup differences were observed for BMI, PSA, and rate of ≥ T3a tumors on pathology. Rate of GG ≥ 2, GG ≥ 3, and GG ≥ 4 showed a significant pattern of increase by PRS quartile (p < 0.001, < 0.001, and 0.039, respectively). With the lowest PRS quartile as reference, higher PRS groups showed sequentially escalating risk for GG ≥ 2 and GG ≥ 3 pathology, with a 4.6-fold rise in GG ≥ 2 (p < 0.001) and 2.0-fold rise in GG ≥ 3 (p < 0.001) for the highest PRS quartiles. Combining PRS with PSA improved prediction of early onset csPCa (AUC 0.759) compared to PRS (AUC 0.627) and PSA alone (AUC 0.736). To conclude, an ethnic-specific PRS was found to predict susceptibility of aggressive PCa in addition to improving detection of csPCa when combined with PSA in early onset populations. PRS may have a role as a risk-stratification model in actual practice. Large scale, multi-ethnic trials are required to validate our results.
Collapse
|
38
|
Varma A, Maharjan J, Garikipati A, Hurtado M, Shokouhi S, Mao Q. Early prediction of prostate cancer risk in younger men using polygenic risk scores and electronic health records. Cancer Med 2023; 12:379-386. [PMID: 35751453 PMCID: PMC9844630 DOI: 10.1002/cam4.4934] [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] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/04/2022] [Accepted: 05/24/2022] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Prostate cancer (PCa) screening is not routinely conducted in men aged 55 and younger, although this age group accounts for more than 10% of cases. Polygenic risk scores (PRSs) and patient data applied toward early prediction of PCa may lead to earlier interventions and increased survival. We have developed machine learning (ML) models to predict PCa risk in men 55 and under using PRSs combined with patient data. METHODS We conducted a retrospective study on 91,106 male patients aged 35-55 using the UK Biobank database. Five gradient boosting models were developed and validated utilizing routine screening data, PRSs, additional clinical data, or combinations of the three. RESULTS Combinations of PRSs and patient data outperformed models that utilized PRS or patient data only, and the highest performing models achieved an area under the receiver operating characteristic curve of 0.788. Our models demonstrated a substantially lower false positive rate (35.4%) in comparison to standard screening using prostate-specific antigen (60%-67%). CONCLUSION This study provides the first preliminary evidence for the use of PRSs with patient data in a ML algorithm for PCa risk prediction in men aged 55 and under for whom screening is not standard practice.
Collapse
Affiliation(s)
| | | | | | | | | | - Qingqing Mao
- Dascena Inc.HoustonTexasUSA
- Montera Inc.San FranciscoCAUSA
| |
Collapse
|
39
|
Pagadala MS, Linscott JA, Talwar JV, Seibert TM, Rose B, Lynch J, Panizzon M, Hauger R, Hansen MH, Sammon JD, Hayn MH, Kader K, Carter H, Ryan ST. PRState: Incorporating genetic ancestry in prostate cancer risk scores for men of African ancestry. BMC Cancer 2022; 22:1289. [PMID: 36494783 PMCID: PMC9733391 DOI: 10.1186/s12885-022-10258-3] [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] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 10/30/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Prostate cancer (PrCa) is one of the most genetically driven solid cancers with heritability estimates as high as 57%. Men of African ancestry are at an increased risk of PrCa; however, current polygenic risk score (PRS) models are based on European ancestry groups and may not be broadly applicable. The objective of this study was to construct an African ancestry-specific PrCa PRS (PRState) and evaluate its performance. METHODS African ancestry group of 4,533 individuals in ELLIPSE consortium was used for discovery of African ancestry-specific PrCa SNPs. PRState was constructed as weighted sum of genotypes and effect sizes from genome-wide association study (GWAS) of PrCa in African ancestry group. Performance was evaluated using ROC-AUC analysis. RESULTS We identified African ancestry-specific PrCa risk loci on chromosomes 3, 8, and 11 and constructed a polygenic risk score (PRS) from 10 African ancestry-specific PrCa risk SNPs, achieving an AUC of 0.61 [0.60-0.63] and 0.65 [0.64-0.67], when combined with age and family history. Performance dropped significantly when using ancestry-mismatched PRS models but remained comparable when using trans-ancestry models. Importantly, we validated the PRState score in the Million Veteran Program (MVP), demonstrating improved prediction of PrCa and metastatic PrCa in individuals of African ancestry. CONCLUSIONS African ancestry-specific PRState improves PrCa prediction in African ancestry groups in ELLIPSE consortium and MVP. This study underscores the need for inclusion of individuals of African ancestry in gene variant discovery to optimize PRSs and identifies African ancestry-specific variants for use in future studies.
Collapse
Affiliation(s)
- Meghana S Pagadala
- Department of Medicine, Division of Medical Genetics, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA, 92093, USA.
- Medical Scientist Training Program, University of California San Diego, La Jolla, CA, USA.
- Biomedical Science Program, University of California, San Diego, La Jolla, CA, USA.
| | | | - James V Talwar
- Bioinformatics and Systems Biology Program, University of California, San Diego, La Jolla, CA, USA
| | - Tyler M Seibert
- Department of Radiation Medicine and Applied Sciences, University of California, San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, La Jolla, CA, USA
- Department of Radiology, University of California San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Brent Rose
- Department of Radiation Medicine and Applied Sciences, University of California, San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, La Jolla, CA, USA
- Department of Urology, University of California San Diego, La Jolla, CA, USA
| | - Julie Lynch
- VA Salt Lake City Healthcare System, Salt Lake City, UT, USA
- School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Matthew Panizzon
- VA San Diego Healthcare System, La Jolla, CA, USA
- Center for Behavioral Genetics of Aging, University of California San Diego, La Jolla, CA, USA
| | - Richard Hauger
- VA San Diego Healthcare System, La Jolla, CA, USA
- Center for Behavioral Genetics of Aging, University of California San Diego, La Jolla, CA, USA
- Center of Excellence for Stress and Mental Health (CESAMH), San Diego Healthcare System, San Diego, CA, USA
| | - Moritz H Hansen
- Division of Urology, Maine Medical Center, Portland, ME, USA
| | - Jesse D Sammon
- Division of Urology, Maine Medical Center, Portland, ME, USA
| | - Matthew H Hayn
- Division of Urology, Maine Medical Center, Portland, ME, USA
| | - Karim Kader
- Department of Urology, University of California San Diego, La Jolla, CA, USA
| | - Hannah Carter
- Department of Medicine, Division of Medical Genetics, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA, 92093, USA
| | - Stephen T Ryan
- Division of Urology, Maine Medical Center, Portland, ME, USA
| |
Collapse
|
40
|
Zaigham S, Gonçalves I, Center RG, Engström G, Sun J. Polygenic scores for low lung function and the future risk of adverse health outcomes. Cardiovasc Diabetol 2022; 21:230. [PMCID: PMC9635172 DOI: 10.1186/s12933-022-01661-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/07/2022] [Indexed: 11/05/2022] Open
Abstract
Aims Reduced lung function and adverse health outcomes are often observed. This study characterizes genetic susceptibility for reduced lung function and risk of developing a range of adverse health outcomes. Methods We studied 27,438 middle-aged adults from the Malmö Diet and Cancer study (MDCS), followed up to 28.8 years. Trait-specific Polygenic scores (PGS) for forced expiratory volume in 1 s (FEV1) and forced vital capacity (FVC) were constructed for each participant using MDCS genetic data and summary statistics from the latest GWAS of lung function. Linear regression models and cox proportional hazards regression models were used to assess associations between adverse health outcomes and lung function-PGS. Results FEV1-PGS and FVC-PGS were significantly associated with mean sBP at baseline after adjustments (FEV1-PGS Q1 (highest PGS = highest lung function): 140.7mmHg vs. Q4: 141.5mmHg, p-value 0.008). A low FVC-PGS was significantly associated with the risk of future diabetic events after adjustments (Q4 vs. Q1 HR: 1.22 (CI 1.12–1.32), p-trend < 0.001) and had added value to risk prediction models for diabetes. Low FEV1-PGS was significantly associated with future coronary events (Q4 vs. Q1 HR: 1.13 (CI: 1.04–1.22), p-trend 0.008). No significant association was found between PGS and sudden cardiac death, chronic kidney disease or all-cause mortality. Results remained largely unchanged in a subgroup of subjects when further adjusted for apolipoproteins. Conclusion Genetic susceptibility for reduced lung function is associated with higher sBP, increased risk of diabetes and to a lesser extent, future coronary events, suggesting etiological roles of lung function on these outcomes. Using PGS, high-risk groups could be early detected to implement early lifestyle changes to mitigate the risk. Supplementary Information The online version contains supplementary material available at 10.1186/s12933-022-01661-y.
Collapse
Affiliation(s)
- Suneela Zaigham
- grid.4514.40000 0001 0930 2361Department of Clinical Sciences Malmö, Lund University, Malmo, Sweden ,grid.8993.b0000 0004 1936 9457Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Isabel Gonçalves
- grid.4514.40000 0001 0930 2361Department of Clinical Sciences Malmö, Lund University, Malmo, Sweden ,grid.411843.b0000 0004 0623 9987Department of Cardiology, Skåne University Hospital, Malmo, Sweden
| | - Regeneron Genetics Center
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals, Tarrytown, NY USA
| | - Gunnar Engström
- grid.4514.40000 0001 0930 2361Department of Clinical Sciences Malmö, Lund University, Malmo, Sweden
| | - Jiangming Sun
- grid.4514.40000 0001 0930 2361Department of Clinical Sciences Malmö, Lund University, Malmo, Sweden
| |
Collapse
|
41
|
Reliability of Ancestry-specific Prostate Cancer Genetic Risk Score in Four Racial and Ethnic Populations. EUR UROL SUPPL 2022; 45:23-30. [DOI: 10.1016/j.euros.2022.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2022] [Indexed: 11/22/2022] Open
|
42
|
Miyahira AK, Soule HR. The 28th Annual Prostate Cancer Foundation Scientific Retreat report. Prostate 2022; 82:1346-1377. [PMID: 35852016 DOI: 10.1002/pros.24409] [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: 06/21/2022] [Accepted: 06/24/2022] [Indexed: 11/07/2022]
Abstract
BACKGROUND The 28th Annual Prostate Cancer Foundation (PCF) Scientific Retreat was held virtually over 4 days, on October 28-29 and November 4-5, 2021. METHODS The Annual PCF Scientific Retreat is a leading global scientific conference that focuses on first-in-field, unpublished, and high-impact basic, translational, and clinical prostate cancer research, as well as research from other fields with high probability for impacting prostate cancer research and patient care. RESULTS Primary areas of research discussed at the 2021 PCF Retreat included: (i) prostate cancer disparities; (ii) prostate cancer survivorship; (iii) next-generation precision medicine; (iv) PSMA theranostics; (v) prostate cancer lineage plasticity; (vi) tumor metabolism as a cancer driver and treatment target; (vii) prostate cancer genetics and polygenic risk scores; (viii) glucocorticoid receptor biology in castration-resistant prostate cancer (CRPC); (ix) therapeutic degraders; (x) new approaches for immunotherapy in prostate cancer; (xi) novel technologies to overcome the suppressive tumor microenvironment; and (xii) real-world evidence and synthetic/virtual control arms. CONCLUSIONS This article provides a summary of the presentations from the 2021 PCF Scientific Retreat. We hope that sharing this knowledge will help to improve the understanding of the current state of research and direct new advances in prostate cancer research and care.
Collapse
Affiliation(s)
- Andrea K Miyahira
- Science Department, Prostate Cancer Foundation, Santa Monica, California, USA
| | - Howard R Soule
- Science Department, Prostate Cancer Foundation, Santa Monica, California, USA
| |
Collapse
|
43
|
Fusar-Poli P, Manchia M, Koutsouleris N, Leslie D, Woopen C, Calkins ME, Dunn M, Tourneau CL, Mannikko M, Mollema T, Oliver D, Rietschel M, Reininghaus EZ, Squassina A, Valmaggia L, Kessing LV, Vieta E, Correll CU, Arango C, Andreassen OA. Ethical considerations for precision psychiatry: A roadmap for research and clinical practice. Eur Neuropsychopharmacol 2022; 63:17-34. [PMID: 36041245 DOI: 10.1016/j.euroneuro.2022.08.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/04/2022] [Accepted: 08/05/2022] [Indexed: 12/14/2022]
Abstract
Precision psychiatry is an emerging field with transformative opportunities for mental health. However, the use of clinical prediction models carries unprecedented ethical challenges, which must be addressed before accessing the potential benefits of precision psychiatry. This critical review covers multidisciplinary areas, including psychiatry, ethics, statistics and machine-learning, healthcare and academia, as well as input from people with lived experience of mental disorders, their family, and carers. We aimed to identify core ethical considerations for precision psychiatry and mitigate concerns by designing a roadmap for research and clinical practice. We identified priorities: learning from somatic medicine; identifying precision psychiatry use cases; enhancing transparency and generalizability; fostering implementation; promoting mental health literacy; communicating risk estimates; data protection and privacy; and fostering the equitable distribution of mental health care. We hope this blueprint will advance research and practice and enable people with mental health problems to benefit from precision psychiatry.
Collapse
Affiliation(s)
- Paolo Fusar-Poli
- Early Psychosis: Interventions and Clinical-Detection (EPIC) Lab, Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; South London and Maudsley NHS Foundation Trust, London, UK; Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy.
| | - Mirko Manchia
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy; Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, Cagliari, Italy; Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada
| | | | | | | | - Monica E Calkins
- Neurodevelopment and Psychosis Section and Lifespan Brain Institute of Penn/CHOP, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, USA
| | - Michael Dunn
- Centre for Biomedical Ethics, Yong Loo Lin School of Medicine, National University of Singapore
| | - Christophe Le Tourneau
- Institut Curie, Department of Drug Development and Innovation (D3i), INSERM U900 Research unit, Paris-Saclay University, France
| | - Miia Mannikko
- European Federation of Associations of Families of People with Mental Illness (EUFAMI), Leuven, Belgium
| | - Tineke Mollema
- Global Alliance of Mental Illness Advocacy Networks-Europe (GAMIAN), Brussels, Belgium
| | - Dominic Oliver
- Early Psychosis: Interventions and Clinical-Detection (EPIC) Lab, Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Eva Z Reininghaus
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | - Alessio Squassina
- Section of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, Italy
| | - Lucia Valmaggia
- South London and Maudsley NHS Foundation Trust, London, UK; Department of Psychology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Department of Psychiatry, KU Leuven, Belgium
| | - Lars Vedel Kessing
- Copenhagen Affective disorder Research Center (CADIC), Psychiatric Center Copenhagen, Denmark; Department of clinical Medicine, University of Copenhagen, Denmark
| | - Eduard Vieta
- Hospital Clinic, Institute of Neuroscience, University of Barcelona, IDIBAPS, CIBERSAM, Barcelona, Catalonia, Spain
| | - Christoph U Correll
- The Zucker Hillside Hospital, Department of Psychiatry, Northwell Health, Glen Oaks, NY, USA; Department of Psychiatry and Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA; Center for Psychiatric Neuroscience; The Feinstein Institutes for Medical Research, Manhasset, NY, USA; Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany
| | - Celso Arango
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañón, Gregorio Marañón; Health Research Institute (IiGSM), School of Medicine, Universidad Complutense de Madrid; Biomedical Research Center for Mental Health (CIBERSAM), Madrid, Spain
| | - Ole A Andreassen
- NORMENT, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | | |
Collapse
|
44
|
Wang XY, Wang LL, Liang SZ, Yang C, Xu L, Yu MC, Wang YX, Dong QJ. Prediction of gastric cancer risk by a polygenic risk score of Helicobacter pylori. World J Gastrointest Oncol 2022; 14:1844-1855. [PMID: 36187384 PMCID: PMC9516638 DOI: 10.4251/wjgo.v14.i9.1844] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/29/2022] [Accepted: 08/15/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Genetic variants of Helicobacter pylori (H. pylori) are involved in gastric cancer occurrence. Single nucleotide polymorphisms (SNPs) of H. pylori that are associated with gastric cancer have been reported. The combined effect of H. pylori SNPs on the risk of gastric cancer remains unclear. AIM To assess the performance of a polygenic risk score (PRS) based on H. pylori SNPs in predicting the risk of gastric cancer. METHODS A total of 15 gastric cancer-associated H. pylori SNPs were selected. The associations between these SNPs and gastric cancer were further validated in 1022 global strains with publicly available genome sequences. The PRS model was established based on the validated SNPs. The performance of the PRS for predicting the risk of gastric cancer was assessed in global strains using quintiles and random forest (RF) methods. The variation in the performance of the PRS among different populations of H. pylori was further examined. RESULTS Analyses of the association between selected SNPs and gastric cancer in the global dataset revealed that the risk allele frequencies of six SNPs were significantly higher in gastric cancer cases than non-gastric cancer cases. The PRS model constructed subsequently with these validated SNPs produced significantly higher scores in gastric cancer. The odds ratio (OR) value for gastric cancer gradually increased from the first to the fifth quintile of PRS, with the fifth quintile having an OR value as high as 9.76 (95% confidence interval: 5.84-16.29). The results of RF analyses indicated that the area under the curve (AUC) value for classifying gastric cancer and non-gastric cancer was 0.75, suggesting that the PRS based on H. pylori SNPs was capable of predicting the risk of gastric cancer. Assessing the performance of the PRS among different H. pylori populations demonstrated that it had good predictive power for cancer risk for hpEurope strains, with an AUC value of 0.78. CONCLUSION The PRS model based on H. pylori SNPs had a good performance for assessment of gastric cancer risk. It would be useful in the prediction of final consequences of the H. pylori infection and beneficial for the management of the infection in clinical settings.
Collapse
Affiliation(s)
- Xiao-Yu Wang
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Li-Li Wang
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Shu-Zhen Liang
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Chao Yang
- The Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200000, China
| | - Lin Xu
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Meng-Chao Yu
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Yi-Xuan Wang
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Quan-Jiang Dong
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong Province, China
| |
Collapse
|
45
|
Kim MS, Naidoo D, Hazra U, Quiver MH, Chen WC, Simonti CN, Kachambwa P, Harlemon M, Agalliu I, Baichoo S, Fernandez P, Hsing AW, Jalloh M, Gueye SM, Niang L, Diop H, Ndoye M, Snyper NY, Adusei B, Mensah JE, Abrahams AOD, Biritwum R, Adjei AA, Adebiyi AO, Shittu O, Ogunbiyi O, Adebayo S, Aisuodionoe-Shadrach OI, Nwegbu MM, Ajibola HO, Oluwole OP, Jamda MA, Singh E, Pentz A, Joffe M, Darst BF, Conti DV, Haiman CA, Spies PV, van der Merwe A, Rohan TE, Jacobson J, Neugut AI, McBride J, Andrews C, Petersen LN, Rebbeck TR, Lachance J. Testing the generalizability of ancestry-specific polygenic risk scores to predict prostate cancer in sub-Saharan Africa. Genome Biol 2022; 23:194. [PMID: 36100952 PMCID: PMC9472407 DOI: 10.1186/s13059-022-02766-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 09/05/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Genome-wide association studies do not always replicate well across populations, limiting the generalizability of polygenic risk scores (PRS). Despite higher incidence and mortality rates of prostate cancer in men of African descent, much of what is known about cancer genetics comes from populations of European descent. To understand how well genetic predictions perform in different populations, we evaluated test characteristics of PRS from three previous studies using data from the UK Biobank and a novel dataset of 1298 prostate cancer cases and 1333 controls from Ghana, Nigeria, Senegal, and South Africa. RESULTS Allele frequency differences cause predicted risks of prostate cancer to vary across populations. However, natural selection is not the primary driver of these differences. Comparing continental datasets, we find that polygenic predictions of case vs. control status are more effective for European individuals (AUC 0.608-0.707, OR 2.37-5.71) than for African individuals (AUC 0.502-0.585, OR 0.95-2.01). Furthermore, PRS that leverage information from African Americans yield modest AUC and odds ratio improvements for sub-Saharan African individuals. These improvements were larger for West Africans than for South Africans. Finally, we find that existing PRS are largely unable to predict whether African individuals develop aggressive forms of prostate cancer, as specified by higher tumor stages or Gleason scores. CONCLUSIONS Genetic predictions of prostate cancer perform poorly if the study sample does not match the ancestry of the original GWAS. PRS built from European GWAS may be inadequate for application in non-European populations and perpetuate existing health disparities.
Collapse
Affiliation(s)
- Michelle S Kim
- School of Biological Sciences, Georgia Institute of Technology, 950 Atlantic Dr, Atlanta, GA, 30332, USA
| | - Daphne Naidoo
- Centre for Proteomic and Genomic Research, Cape Town, South Africa
| | - Ujani Hazra
- School of Biological Sciences, Georgia Institute of Technology, 950 Atlantic Dr, Atlanta, GA, 30332, USA
| | - Melanie H Quiver
- School of Biological Sciences, Georgia Institute of Technology, 950 Atlantic Dr, Atlanta, GA, 30332, USA
| | - Wenlong C Chen
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,National Cancer Registry, National Health Laboratory Service, Johannesburg, South Africa
| | - Corinne N Simonti
- School of Biological Sciences, Georgia Institute of Technology, 950 Atlantic Dr, Atlanta, GA, 30332, USA
| | | | - Maxine Harlemon
- School of Biological Sciences, Georgia Institute of Technology, 950 Atlantic Dr, Atlanta, GA, 30332, USA
| | - Ilir Agalliu
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Pedro Fernandez
- Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Ann W Hsing
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | | | | | - Lamine Niang
- Universite Cheikh Anta Diop de Dakar, Dakar, Senegal
| | | | - Medina Ndoye
- Universite Cheikh Anta Diop de Dakar, Dakar, Senegal
| | | | | | - James E Mensah
- Korle-Bu Teaching Hospital and University of Ghana Medical School, Accra, Ghana
| | - Afua O D Abrahams
- Korle-Bu Teaching Hospital and University of Ghana Medical School, Accra, Ghana
| | - Richard Biritwum
- Korle-Bu Teaching Hospital and University of Ghana Medical School, Accra, Ghana
| | - Andrew A Adjei
- Department of Pathology, University of Ghana Medical School, Accra, Ghana
| | | | | | | | - Sikiru Adebayo
- College of Medicine, University of Ibadan, Ibadan, Nigeria
| | | | - Maxwell M Nwegbu
- College of Health Sciences, University of Abuja and University of Abuja Teaching Hospital, Abuja, Nigeria
| | - Hafees O Ajibola
- College of Health Sciences, University of Abuja and University of Abuja Teaching Hospital, Abuja, Nigeria
| | - Olabode P Oluwole
- College of Health Sciences, University of Abuja and University of Abuja Teaching Hospital, Abuja, Nigeria
| | - Mustapha A Jamda
- College of Health Sciences, University of Abuja and University of Abuja Teaching Hospital, Abuja, Nigeria
| | - Elvira Singh
- National Cancer Registry, National Health Laboratory Service, Johannesburg, South Africa
| | - Audrey Pentz
- Non-Communicable Diseases Research Division, Wits Health Consortium (PTY) Ltd, Johannesburg, South Africa
| | - Maureen Joffe
- Non-Communicable Diseases Research Division, Wits Health Consortium (PTY) Ltd, Johannesburg, South Africa.,MRC Developmental Pathways to Health Research Unit, Department of Pediatrics, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Burcu F Darst
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - David V Conti
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Christopher A Haiman
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Petrus V Spies
- Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - André van der Merwe
- Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Thomas E Rohan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Judith Jacobson
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Alfred I Neugut
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Jo McBride
- Centre for Proteomic and Genomic Research, Cape Town, South Africa
| | | | | | - Timothy R Rebbeck
- Dana-Farber Cancer Institute, Boston, MA, USA.,Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Joseph Lachance
- School of Biological Sciences, Georgia Institute of Technology, 950 Atlantic Dr, Atlanta, GA, 30332, USA.
| |
Collapse
|
46
|
Zhao L, Luo T, Jiang J, Wu J, Zhang X. Eight gene mutation-based polygenic hazard score as a potential predictor for immune checkpoint inhibitor therapy outcome in metastatic melanoma. Front Mol Biosci 2022; 9:1001792. [PMID: 36120536 PMCID: PMC9478752 DOI: 10.3389/fmolb.2022.1001792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 08/10/2022] [Indexed: 12/24/2022] Open
Abstract
Background: Immune checkpoint inhibitor (ICI) therapies have revolutionized the treatment of metastatic cutaneous melanoma, but have only benefitted a subset of them. Gene mutations were reported to impact the ICI therapy outcomes in metastatic melanoma but have not been fully investigated. Hence, we systematically analyzed the impact of cancer-related gene mutations on the clinical outcome in metastatic melanoma patients who underwent ICI therapies.Methods: Publicly available discovery and validation cohorts (312 patients and 110 patients respectively, all the patients received ICI therapies) were included in this study. Cox proportional hazards regression analysis was used to assess the association of 468 cancer-related gene mutations with overall survival (OS) in the discovery cohort, and the polygenic hazard score (PHS) was constructed subsequently, and validated in the validation cohort. The Tumor Immune Estimation Resource (TIMER) online tools, which are based on The Cancer Genome Atlas database, were used to analyze the impact of gene mutations on tumor-infiltrated immune cells in melanoma samples.Results: We found eight gene mutations that were significantly associated with the overall survival (BAP1, CARD11, IGF1R, KMT2D, PTPRD, PTPRT, ROS1, and TERT, p < 0.05, mutation frequency >0.05). The PHS, which was based on these genes, was found to effectively discriminate the subset which benefited most from ICI therapies (HR = 1·54, 95%CI, 1.25–1.95; p < 0.001). After adjusting with age, sex, ICI regimes, and tumor mutation burden (TMB), we found that PHS was an independent predictor for the outcome of ICI therapies (adjusted HR = 1.84, 95%CI, 1.22–2.79; p = 0.004). The PHS was validated in the validation cohort (log-Rank p = 0.038). Further research found that CARD11 and PTPRD mutations were significantly associated with more tumor-infiltrated immune cells in melanoma samples.Conclusion: For the first time, we have shown that PHS can independently and effectively predict the ICI therapy outcome in metastatic melanoma, which once validated by larger research, may help the decision-making process in melanoma.
Collapse
Affiliation(s)
- Liqin Zhao
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Gastrointestinal Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ting Luo
- Department of Oncology, Chengdu First People’s Hospital, Chengdu, China
| | - Jinling Jiang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junwei Wu
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaowei Zhang
- Department of Gastrointestinal Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- *Correspondence: Xiaowei Zhang,
| |
Collapse
|
47
|
Siltari A, Lönnerbro R, Pang K, Shiranov K, Asiimwe A, Evans-Axelsson S, Franks B, Kiran A, Murtola TJ, Schalken J, Steinbeisser C, Bjartell A, Auvinen A, Smith E, N'Dow J, Plass K, Ribal M, Mottet N, Moris L, Lardas M, Van den Broeck T, Willemse PP, Gandaglia G, Campi R, Greco I, Gacci M, Serni S, Briganti A, Crosti D, Meoni M, Garzonio R, Bangma R, Roobol M, Remmers S, Tilki D, Visakorpi T, Talala K, Tammela T, van Hemelrijck M, Bayer K, Lejeune S, Taxiarchopoulou G, van Diggelen F, Senthilkumar K, Schutte S, Byrne S, Fialho L, Cardone A, Gono P, De Vetter M, Ceke K, De Meulder B, Auffray C, Balaur IA, Taibi N, Power S, Kermani NZ, van Bochove K, Cavelaars M, Moinat M, Voss E, Bernini C, Horgan D, Fullwood L, Holtorf M, Lancet D, Bernstein G, Omar I, MacLennan S, Maclennan S, Healey J, Huber J, Wirth M, Froehner M, Brenner B, Borkowetz A, Thomas C, Horn F, Reiche K, Kreux M, Josefsson A, Tandefekt DG, Hugosson J, Huisman H, Hofmacher T, Lindgren P, Andersson E, Fridhammar A, Vizcaya D, Verholen F, Zong J, Butler-Ransohoff JE, Williamson T, Chandrawansa K, Dlamini D, waldeck R, Molnar M, Bruno A, Herrera R, Jiang S, et alSiltari A, Lönnerbro R, Pang K, Shiranov K, Asiimwe A, Evans-Axelsson S, Franks B, Kiran A, Murtola TJ, Schalken J, Steinbeisser C, Bjartell A, Auvinen A, Smith E, N'Dow J, Plass K, Ribal M, Mottet N, Moris L, Lardas M, Van den Broeck T, Willemse PP, Gandaglia G, Campi R, Greco I, Gacci M, Serni S, Briganti A, Crosti D, Meoni M, Garzonio R, Bangma R, Roobol M, Remmers S, Tilki D, Visakorpi T, Talala K, Tammela T, van Hemelrijck M, Bayer K, Lejeune S, Taxiarchopoulou G, van Diggelen F, Senthilkumar K, Schutte S, Byrne S, Fialho L, Cardone A, Gono P, De Vetter M, Ceke K, De Meulder B, Auffray C, Balaur IA, Taibi N, Power S, Kermani NZ, van Bochove K, Cavelaars M, Moinat M, Voss E, Bernini C, Horgan D, Fullwood L, Holtorf M, Lancet D, Bernstein G, Omar I, MacLennan S, Maclennan S, Healey J, Huber J, Wirth M, Froehner M, Brenner B, Borkowetz A, Thomas C, Horn F, Reiche K, Kreux M, Josefsson A, Tandefekt DG, Hugosson J, Huisman H, Hofmacher T, Lindgren P, Andersson E, Fridhammar A, Vizcaya D, Verholen F, Zong J, Butler-Ransohoff JE, Williamson T, Chandrawansa K, Dlamini D, waldeck R, Molnar M, Bruno A, Herrera R, Jiang S, Nevedomskaya E, Fatoba S, Constantinovici N, Maass M, Torremante P, Voss M, Devecseri Z, Cuperus G, Abott T, Dau C, Papineni K, Wang-Silvanto J, Hass S, Snijder R, Doye V, Wang X, Garnham A, Lambrecht M, Wolfinger R, Rogiers S, Servan A, Lefresne F, Caseriego J, Samir M, Lawson J, Pacoe K, Robinson P, Jaton B, Bakkard D, Turunen H, Kilkku O, Pohjanjousi P, Voima O, Nevalaita L, Reich C, Araujo S, Longden-Chapman E, Burke D, Agapow P, Derkits S, Licour M, McCrea C, Payne S, Yong A, Thompson L, Lujan F, Bussmann M, Köhler I. How well do polygenic risk scores identify men at high risk for prostate cancer? Systematic review and meta-analysis. Clin Genitourin Cancer 2022; 21:316.e1-316.e11. [PMID: 36243664 DOI: 10.1016/j.clgc.2022.09.006] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/01/2022] [Accepted: 09/06/2022] [Indexed: 11/27/2022]
Abstract
OBJECTIVES Genome-wide association studies have revealed over 200 genetic susceptibility loci for prostate cancer (PCa). By combining them, polygenic risk scores (PRS) can be generated to predict risk of PCa. We summarize the published evidence and conduct meta-analyses of PRS as a predictor of PCa risk in Caucasian men. PATIENTS AND METHODS Data were extracted from 59 studies, with 16 studies including 17 separate analyses used in the main meta-analysis with a total of 20,786 cases and 69,106 controls identified through a systematic search of ten databases. Random effects meta-analysis was used to obtain pooled estimates of area under the receiver-operating characteristic curve (AUC). Meta-regression was used to assess the impact of number of single-nucleotide polymorphisms (SNPs) incorporated in PRS on AUC. Heterogeneity is expressed as I2 scores. Publication bias was evaluated using funnel plots and Egger tests. RESULTS The ability of PRS to identify men with PCa was modest (pooled AUC 0.63, 95% CI 0.62-0.64) with moderate consistency (I2 64%). Combining PRS with clinical variables increased the pooled AUC to 0.74 (0.68-0.81). Meta-regression showed only negligible increase in AUC for adding incremental SNPs. Despite moderate heterogeneity, publication bias was not evident. CONCLUSION Typically, PRS accuracy is comparable to PSA or family history with a pooled AUC value 0.63 indicating mediocre performance for PRS alone.
Collapse
|
48
|
Inherited risk assessment and its clinical utility for predicting prostate cancer from diagnostic prostate biopsies. Prostate Cancer Prostatic Dis 2022; 25:422-430. [PMID: 35347252 DOI: 10.1038/s41391-021-00458-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 08/25/2021] [Accepted: 09/10/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Many studies on prostate cancer (PCa) germline variants have been published in the last 15 years. This review critically assesses their clinical validity and explores their utility in prediction of PCa detection rates from prostate biopsy. METHODS An integrative review was performed to (1) critically synthesize findings on PCa germline studies from published papers since 2016, including risk-associated single nucleotide polymorphisms (SNPs), polygenic risk score methods such as genetic risk score (GRS), and rare pathogenic mutations (RPMs); (2) exemplify the findings in a large population-based cohort from the UK Biobank (UKB); (3) identify gaps for implementing inherited risk assessment in clinic based on experience from a healthcare system; (4) evaluate available GRS data on their clinical utility in predicting PCa detection rates from prostate biopsies; and (5) describe a prospective germline-based biopsy trial to address existing gaps. RESULTS SNP-based GRS and RPMs in four genes (HOXB13, BRCA2, ATM, and CHEK2) were significantly and consistently associated with PCa risk in large well-designed studies. In the UKB, positive family history, RPMs in the four implicated genes, and a high GRS (>1.5) identified 8.12%, 1.61%, and 17.38% of men to be at elevated PCa risk, respectively, with hazard ratios of 1.84, 2.74, and 2.39, respectively. Additionally, the performance of GRS for predicting PCa detection rate on prostate biopsy was consistently supported in several retrospective analyses of transrectal ultrasound (TRUS)-biopsy cohorts. Prospective studies evaluating the performance of all three inherited measures in predicting PCa detection rate from contemporary multiparametric MRI (mpMRI)-based biopsy are lacking. A multicenter germline-based biopsy trial to address these gaps is warranted. CONCLUSIONS The complementary performance of three inherited risk measures in PCa risk stratification is consistently supported. Their clinical utility in predicting PCa detection rate, if confirmed in prospective clinical trials, may improve current decision-making for prostate biopsy.
Collapse
|
49
|
Multiple primary cancers in men with sporadic or familial prostate cancer: Its clinical implications. Urol Oncol 2022; 40:489.e1-489.e7. [DOI: 10.1016/j.urolonc.2022.07.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 06/08/2022] [Accepted: 07/19/2022] [Indexed: 11/20/2022]
|
50
|
Arenas-Gallo C, Owiredu J, Weinstein I, Lewicki P, Basourakos SP, Vince R, Al Hussein Al Awamlh B, Schumacher FR, Spratt DE, Barbieri CE, Shoag JE. Race and prostate cancer: genomic landscape. Nat Rev Urol 2022; 19:547-561. [PMID: 35945369 DOI: 10.1038/s41585-022-00622-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/23/2022] [Indexed: 11/09/2022]
Abstract
In the past 20 years, new insights into the genomic pathogenesis of prostate cancer have been provided. Large-scale integrative genomics approaches enabled researchers to characterize the genetic and epigenetic landscape of prostate cancer and to define different molecular subclasses based on the combination of genetic alterations, gene expression patterns and methylation profiles. Several molecular drivers of prostate cancer have been identified, some of which are different in men of different races. However, the extent to which genomics can explain racial disparities in prostate cancer outcomes is unclear. Future collaborative genomic studies overcoming the underrepresentation of non-white patients and other minority populations are essential.
Collapse
Affiliation(s)
- Camilo Arenas-Gallo
- Department of Urology, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Jude Owiredu
- Department of Urology, NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, USA
| | - Ilon Weinstein
- Department of Urology, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Patrick Lewicki
- Department of Urology, NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, USA
| | - Spyridon P Basourakos
- Department of Urology, NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, USA
| | - Randy Vince
- Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - Bashir Al Hussein Al Awamlh
- Department of Urology, NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, USA.,Department of Urology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Fredrick R Schumacher
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Daniel E Spratt
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Christopher E Barbieri
- Department of Urology, NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, USA
| | - Jonathan E Shoag
- Department of Urology, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA. .,Department of Urology, NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, USA. .,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|