Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Trans-ancestry genome-wide association meta-analysis of prostate cancer identifies new susceptibility loci and informs genetic risk prediction

A Publisher Correction to this article was published on 20 January 2021

This article has been updated

Abstract

Prostate cancer is a highly heritable disease with large disparities in incidence rates across ancestry populations. We conducted a multiancestry meta-analysis of prostate cancer genome-wide association studies (107,247 cases and 127,006 controls) and identified 86 new genetic risk variants independently associated with prostate cancer risk, bringing the total to 269 known risk variants. The top genetic risk score (GRS) decile was associated with odds ratios that ranged from 5.06 (95% confidence interval (CI), 4.84–5.29) for men of European ancestry to 3.74 (95% CI, 3.36–4.17) for men of African ancestry. Men of African ancestry were estimated to have a mean GRS that was 2.18-times higher (95% CI, 2.14–2.22), and men of East Asian ancestry 0.73-times lower (95% CI, 0.71–0.76), than men of European ancestry. These findings support the role of germline variation contributing to population differences in prostate cancer risk, with the GRS offering an approach for personalized risk prediction.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: OR for prostate cancer by GRS category stratified by age.
Fig. 2: Comparison of prostate cancer GRS distributions for controls.
Fig. 3: Absolute risks of prostate cancer by GRS category.

Similar content being viewed by others

Data availability

The full summary statistics resulting from this investigation are available through dbGaP under accession code phs001120.v2.p1. The genotype data and relevant covariate information (ancestry, country, principal components and so on) used in this study are deposited in dbGaP under accession codes phs001391.v1.p1, phs000306.v4.p1, phs001120.v1.p1, phs001221.v1.p1, phs000812.v1.p1 and phs000838.v1.p1. Publicly available data described in this manuscript can be found from the following websites: 1000 Genomes Project (https://www.internationalgenome.org/); SEER (https://seer.cancer.gov/); National Center for Health Statistics, CDC (https://www.cdc.gov/nchs/index.htm); Cistrome Data Browser (http://cistrome.org/db/); GTEx (https://gtexportal.org/home/datasets); and TCGA (https://portal.gdc.cancer.gov/).

Code availability

Imputation was performed using IMPUTE2, MACH 1.0, Minimac3 and Minimac4. Association testing was performed using PLINK 1.07, SNPtest v.2.5.2, and R v.3.5. Meta-analyses were conducted using METAL v.2011-03-25 and fine-mapping with JAM. Other analyses were performed with PriorityPruner v.0.1.4, RFMix v.1.0.2 and wANNOVAR (accessed 21 April 2020). Custom code modifying the JAM approach was developed for these analyses and is available on GitHub (https://github.com/USCmec/Conti_NatGen_2020). Code for analyses using other indicated software is readily available from the websites of the corresponding software.

Change history

  • 20 January 2021

    A Correction to this paper has been published: https://doi.org/10.1038/s41588-021-00786-2.

References

  1. US Cancer Statistics Working Group (June 2019). U.S. Cancer Statistics Data Visualizations Tool, based on November 2018 submission data (1999–2016) (US Department of Health and Human Services, Centers for Disease Control and Preventions and National Cancer Institute, accessed 1 September 2019); www.cdc.gov/cancer/dataviz

  2. Mucci, L. A. et al. Familial risk and heritability of cancer among twins in nordic countries. JAMA 315, 68–76 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Freedman, M. L. et al. Admixture mapping identifies 8q24 as a prostate cancer risk locus in African-American men. Proc. Natl Acad. Sci. USA 103, 14068–14073 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Al Olama, A. A. et al. A meta-analysis of 87,040 individuals identifies 23 new susceptibility loci for prostate cancer. Nat. Genet. 46, 1103–1109 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Amundadottir, L. T. et al. A common variant associated with prostate cancer in European and African populations. Nat. Genet. 38, 652–658 (2006).

    CAS  PubMed  Google Scholar 

  6. Conti, D. V. et al. Two novel susceptibility loci for prostate cancer in men of African ancestry. J. Natl. Cancer Inst. 109, djx084 (2017).

    PubMed Central  Google Scholar 

  7. Dadaev, T. et al. Fine-mapping of prostate cancer susceptibility loci in a large meta-analysis identifies candidate causal variants. Nat. Commun. 9, 2256 (2018).

    PubMed  PubMed Central  Google Scholar 

  8. Eeles, R. A. et al. Identification of 23 new prostate cancer susceptibility loci using the iCOGS custom genotyping array. Nat. Genet. 45, 385–391 (2013). 391e1-2.

    CAS  PubMed  Google Scholar 

  9. Gudmundsson, J. et al. A study based on whole-genome sequencing yields a rare variant at 8q24 associated with prostate cancer. Nat. Genet. 44, 1326–1329 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Gudmundsson, J. et al. Common sequence variants on 2p15 and Xp11.22 confer susceptibility to prostate cancer. Nat. Genet. 40, 281–283 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Hoffmann, T. J. et al. A large multiethnic genome-wide association study of prostate cancer identifies novel risk variants and substantial ethnic differences. Cancer Discov. 5, 878–891 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Schumacher, F. R. et al. Association analyses of more than 140,000 men identify 63 new prostate cancer susceptibility loci. Nat. Genet. 50, 928–936 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Takata, R. et al. Genome-wide association study identifies five new susceptibility loci for prostate cancer in the Japanese population. Nat. Genet. 42, 751–754 (2010).

    CAS  PubMed  Google Scholar 

  14. Wang, M. et al. Large-scale association analysis in Asians identifies new susceptibility loci for prostate cancer. Nat. Commun. 6, 8469 (2015).

    CAS  PubMed  Google Scholar 

  15. 1000 Genomes Project Consortium et al. A global reference for human genetic variation. Nature 526, 68–74 (2015).

    Google Scholar 

  16. Kicinski, M., Vangronsveld, J. & Nawrot, T. S. An epidemiological reappraisal of the familial aggregation of prostate cancer: a meta-analysis. PLoS ONE 6, e27130 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Bode, A. M. & Dong, Z. Post-translational modification of p53 in tumorigenesis. Nat. Rev. Cancer 4, 793–805 (2004).

    CAS  PubMed  Google Scholar 

  18. Dong, X. et al. Mutations in CHEK2 associated with prostate cancer risk. Am. J. Hum. Genet. 72, 270–280 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Dowling, C. R. & Risbridger, G. P. The role of inhibins and activins in prostate cancer pathogenesis. Endocr. Relat. Cancer 7, 243–256 (2000).

    CAS  PubMed  Google Scholar 

  20. Lambert, S. A. et al. The human transcription factors. Cell 172, 650–665 (2018).

    CAS  PubMed  Google Scholar 

  21. O’Hurley, G. et al. Analysis of the human prostate-specific proteome defined by transcriptomics and antibody-based profiling identifies TMEM79 and ACOXL as two putative, diagnostic markers in prostate cancer. PLoS ONE 10, e0133449 (2015).

    PubMed  PubMed Central  Google Scholar 

  22. Uhlen, M. et al. Towards a knowledge-based human protein atlas. Nat. Biotechnol. 28, 1248–1250 (2010).

    CAS  PubMed  Google Scholar 

  23. Zaitlen, N., Pasaniuc, B., Gur, T., Ziv, E. & Halperin, E. Leveraging genetic variability across populations for the identification of causal variants. Am. J. Hum. Genet. 86, 23–33 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Duncan, L. et al. Analysis of polygenic risk score usage and performance in diverse human populations. Nat. Commun. 10, 3328 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Moyer, V. A. & Force, U. S. P. S. T. Screening for prostate cancer: U.S. Preventive Services Task Force recommendation statement. Ann. Intern. Med. 157, 120–134 (2012).

    PubMed  Google Scholar 

  26. Li, Y., Willer, C. J., Ding, J., Scheet, P. & Abecasis, G. R. MaCH: using sequence and genotype data to estimate haplotypes and unobserved genotypes. Genet. Epidemiol. 34, 816–834 (2010).

    PubMed  PubMed Central  Google Scholar 

  27. Das, S. et al. Next-generation genotype imputation service and methods. Nat. Genet. 48, 1284–1287 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Howie, B. N., Donnelly, P. & Marchini, J. A flexible and accurate genotype imputation method for the next generation of genome-wide association studies. PLoS Genet. 5, e1000529 (2009).

    PubMed  PubMed Central  Google Scholar 

  29. Bycroft, C. et al. The UK Biobank resource with deep phenotyping and genomic data. Nature 562, 203–209 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Purcell, S. et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am. J. Hum. Genet. 81, 559–575 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Marchini, J., Howie, B., Myers, S., McVean, G. & Donnelly, P. A new multipoint method for genome-wide association studies by imputation of genotypes. Nat. Genet. 39, 906–913 (2007).

    CAS  PubMed  Google Scholar 

  32. Willer, C. J., Li, Y. & Abecasis, G. R. METAL: fast and efficient meta-analysis of genomewide association scans. Bioinformatics 26, 2190–2191 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Newcombe, P. J., Conti, D. V. & Richardson, S. JAM: a scalable Bayesian framework for joint analysis of marginal SNP effects. Genet. Epidemiol. 40, 188–201 (2016).

    PubMed  PubMed Central  Google Scholar 

  34. Ewing, C. M. et al. Germline mutations in HOXB13 and prostate-cancer risk. N. Engl. J. Med. 366, 141–149 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Seppala, E. H. et al. CHEK2 variants associate with hereditary prostate cancer. Br. J. Cancer 89, 1966–1970 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Zhong, H. & Prentice, R. L. Bias-reduced estimators and confidence intervals for odds ratios in genome-wide association studies. Biostatistics 9, 621–634 (2008).

    PubMed  PubMed Central  Google Scholar 

  37. Zhong, H. & Prentice, R. L. Correcting “winner’s curse” in odds ratios from genomewide association findings for major complex human diseases. Genet. Epidemiol. 34, 78–91 (2010).

    PubMed  PubMed Central  Google Scholar 

  38. Pencina, M. J., D’Agostino, R. B. Sr. & Steyerberg, E. W. Extensions of net reclassification improvement calculations to measure usefulness of new biomarkers. Stat. Med. 30, 11–21 (2011).

    PubMed  Google Scholar 

  39. Maples, B. K., Gravel, S., Kenny, E. E. & Bustamante, C. D. RFMix: a discriminative modeling approach for rapid and robust local-ancestry inference. Am. J. Hum. Genet. 93, 278–288 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Amin Al Olama, A. et al. Risk analysis of prostate cancer in PRACTICAL, a multinational consortium, using 25 known prostate cancer susceptibility loci. Cancer Epidemiol. Biomarkers Prev. 24, 1121–1129 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Antoniou, A. C. et al. Common breast cancer susceptibility alleles and the risk of breast cancer for BRCA1 and BRCA2 mutation carriers: implications for risk prediction. Cancer Res. 70, 9742–9754 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Antoniou, A. C. et al. Evidence for further breast cancer susceptibility genes in addition to BRCA1 and BRCA2 in a population-based study. Genet. Epidemiol. 21, 1–18 (2001).

    CAS  PubMed  Google Scholar 

  43. Kuchenbaecker, K. B. et al. Evaluation of polygenic risk scores for breast and ovarian cancer risk prediction in BRCA1 and BRCA2 mutation carriers. J. Natl. Cancer Inst. 109, djw302 (2017).

    PubMed Central  Google Scholar 

  44. Wang, K. et al. Interpretation of association signals and identification of causal variants from genome-wide association studies. Am. J. Hum. Genet. 86, 730–742 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Witte, J. S., Visscher, P. M. & Wray, N. R. The contribution of genetic variants to disease depends on the ruler. Nat. Rev. Genet. 15, 765–776 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Chang, X. & Wang, K. wANNOVAR: annotating genetic variants for personal genomes via the web. J. Med. Genet. 49, 433–436 (2012).

    PubMed  Google Scholar 

  47. Mei, S. et al. Cistrome Data Browser: a data portal for ChIP-seq and chromatin accessibility data in human and mouse. Nucleic Acids Res. 45, D658–D662 (2017).

    CAS  PubMed  Google Scholar 

  48. Lawrence, M., Gentleman, R. & Carey, V. rtracklayer: an R package for interfacing with genome browsers. Bioinformatics 25, 1841–1842 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. GTEx Consortium. The genotype-tissue expression (GTEx) project. Nat. Genet. 45, 580–585 (2013).

    Google Scholar 

  50. Thibodeau, S. N. et al. Identification of candidate genes for prostate cancer-risk SNPs utilizing a normal prostate tissue eQTL data set. Nat. Commun. 6, 8653 (2015).

    CAS  PubMed  Google Scholar 

  51. Ongen, H., Buil, A., Brown, A. A., Dermitzakis, E. T. & Delaneau, O. Fast and efficient QTL mapper for thousands of molecular phenotypes. Bioinformatics 32, 1479–1485 (2016).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This project was support by US National Institutes of Health grants no. U19CA148537 (C.A.H.), no. U01CA194393 (S. Lindstrom) and no. K99CA246063 (B.F. Darst). We acknowledge the ARCS Foundation, Inc., Los Angeles Chapter, for their generous support of L.C.M. through the Margaret Kirsten Ponty Fellowship and B.F. Darst through the John and Edith Leonis Family Foundation. This research has been conducted using the UK Biobank Resource under application no. 42195. A full description of funding and acknowledgements for each of the contributing studies can be found in the Supplementary Information.

Author information

Authors and Affiliations

Authors

Contributions

D.V.C., R.A.E., Z.K.-J. and C.A.H. contributed to study conception, and D.V.C., B.F. Darst, R.A.E., Z.K.-J. and C.A.H. contributed to interpretation and wrote the manuscript. E.J.S. performed a literature search. M.B., T.D., S.B. and X.S. provided data management and bioinformatics support. D.V.C., L.C.M., B.F. Darst, E.J.S., T.D., Z.K.-J. and C.A.H. contributed to data analysis and interpretation. All authors contributed data to the study, and revised, critically reviewed and approved the final version of the manuscript: D.V.C., B.F. Darst, L.C.M., E.J.S., X.S., A.C., F.R.S., A.A.A.O., S.B., T.D., M.N.B., A.S., T.J.H., A.T., K. Matsuda, Y.M., M.F., K. Muir, A. Lophatananon, P.W., L.L.M., L.R.W., V.L.S., S.M.G., B.D.C., J.S., T.L.J.T., C. Sipeky, A.A., G.G.G., M.C. Southey, R.J.M., C.C., D.W., J. Lubiński, D.E.N., J.L.D., F.C.H., R.M.M., B.G.N., S.F.N., M.W., S.E.B., M.A.R., P.I., J.B., S.C., L. Moya, L.H., J.A.C., W.T., G.P.R., H.G., M.A., R.S., M.E., T.N., N.P., A.M.D., M. Ghoussaini, R.C.T., T.J.K., E.R., J.Y.P., T.A.S., H.-Y.L., D.A., S.J.W., L.A.M., E.G., S. Lindstrom, P.K., D.J.H., K.L.P., C.T., C.M.T., P.J.G., I.M.T., R.J.H., N.E.F., A.F., M.-É.P., J.L.S., E.A.O., M.S.G., S.K., L.E.B.F., M. Stampfer, A.W., N.H., G.L.A., R.N.H., M.J.M., K.D.S., M.B., W.J.B., W.Z., E.D.Y., J.E.M., Y.-J.L., H.-W.Z., N.F., X.M., Y.W., S.-C.Z., Z.S., S.N.T., S.K.M., D.J.S., C.M.L.W., N.B., G.B., C.M., T.S., M.L., A.S.K., B.F. Drake, O.C., G.C.-T., F.M., T.T., Y.A.K., E.M.J., E.M.G., L. Maehle, K.-T.K., S.A.I., M.C. Stern, A.V., A.G.-C., L.F., B.S.R., S.L.K., H.O., M.R.T., P.P., A.B., S.W., A. Lubwama, J.T.B., E.T.H.F., J.M., J.A.T., M.K., J. Llorca, G.C.-V., L.C.-A., C.C.T., C.D.H., S.S.S., L. Multigner, P.B., L.B., R.K., C. Slavov, V.M., R.J.L., B.W., H.B., K.C., B.H., K.-U.S., E.A.K., A.W.H., R.A.K., A.B.M., C.J.L., J.K., S.L.N., L.S., Y.C.D., W.B.I., B.N., A.J.M.H., J.C., H.P., A.M., K.D.R., G.D.M., P.O., J.X., A.R., J. Lim, S.-H.T., L.F.N., D.W.L., J.H.F., C.N.-D., B.A.R., M. Ghoussaini, D.L., T.K., N.U., S. Singhal, M.P., F.C., S.J., T.V.B., M.G.-D., J.E.C., M.E.M., S. Larkin, P.A.T., C.A.-H., W.S.B., M.C.A., D.C.C., S. Srivastava, J.C.C., G.P., G.C., M.J.R., G.J., R.H.N.S., J.J.H., M. Sanderson, R.V., R.M.-C., M.T., N.M., S.I.B., S.K.V.D.E., D.F.E., S.J.C., M.B.C., F.W., H.N., J.S.W., R.A.E., Z.K.-J. and C.A.H. C.A.H. and R.A.E. had full access to all the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis.

Corresponding author

Correspondence to Christopher A. Haiman.

Ethics declarations

Competing interests

R.A.E. reports the following disclosures: (1) GU-ASCO meeting in San Francisco (January 2016)—received US$500 honorarium as speaker; (2) RMH FR meeting (November 2017)—received support from Janssen and £1,100 honorarium as speaker; (3) University of Chicago invited talk (May 2018)—received US$1,000 honorarium as speaker; (4) EUR 200 education honorarium paid by Bayer & Ipsen to attend GU Connect ‘Treatment sequencing for mCRPC patients within the changing landscape of mHSPC’ at a venue at ESMO, Barcelona (September 2019); and (5) Prostate Dx Advisory Panel—Member of External Expert Committee (June 2020), 3 hours, £900. The remaining authors declare no competing interests.

Additional information

Peer review information Nature Genetics thanks Robert Bristow and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Effect comparisons of the 269 prostate cancer risk variants between younger (age ≤ 55) and older (age > 55) men of European and African ancestry.

Variants above the identity line have larger effects in younger men, and variants below the identity line have larger effects in older men. Blue dots indicate effect differences with an unadjusted P-value < 0.05. 188/269 (69.9%) of tested variants have larger effects in younger vs. older men and 31/269 (11.5%) of tested variants have larger effects in younger vs. older men at a P-value < 0.05 threshold. All statistical tests were two-sided. Results presented in the figure are also provided in Supplementary Table 8. SE: standard error.

Extended Data Fig. 2 Effect correlations of the 269 prostate cancer risk variants between populations.

Effects and RAF are compared between European (EUR) ancestry men and a) African (AFR) ancestry men, b) East Asian (EAS) ancestry men, and c) Hispanic (HIS) men. Figure is annotated to show risk allele frequency (RAF) differences between Europeans and non-Europeans for each variant. SE: standard error.

Extended Data Fig. 3 Discriminative ability and highest GRS decile odds ratio of the multiethnic genome-wide GRS upon iteratively adding each variant to the GRS model.

a) European ancestry men from the UK Biobank and b) African ancestry men from the California Uganda (CA UG) study. Variants are sorted first within the 269-variant genetic risk score (GRS) then for other genome-wide variants by the multiethnic genome-wide association study (GWAS) meta-analysis P-values (with four P-value thresholds indicated by dotted vertical lines), and GRS weights are based on multiancestry GWAS meta-analysis results. Black lines represent the area under the curve (AUC) and correspond to the left y-axis, while blue lines represent the 90-100% GRS odds ratio (OR; relative to 40-60% GRS) and correspond to the right y-axis. All statistical tests were two-sided. PCs: principal components.

Extended Data Fig. 4 Discriminative ability and highest GRS decile odds ratio of the African ancestry genome-wide GRS upon iteratively adding each variant to the GRS model.

a) European ancestry men from the UK Biobank and b) African ancestry men from the California Uganda (CA UG) study. Variants are sorted first within the 269-variant genetic risk score (GRS) then for other genome-wide variants by the African ancestry genome-wide association study (GWAS) meta-analysis P-values (with four P-value thresholds indicated by dotted vertical lines), and GRS weights are based on African ancestry GWAS meta-analysis results. Black lines represent the area under the curve (AUC) and correspond to the left y-axis, while blue lines represent the 90-100% GRS odds ratio (OR; relative to 40-60% GRS) and correspond to the right y-axis. All statistical tests were two-sided. PCs: principal components.

Extended Data Fig. 5 Distribution of age at prostate cancer diagnosis by GRS category and population.

Differences between populations reflect sampling differences rather than population differences in age at diagnosis. SE: standard deviation, GRS: genetic risk score.

Extended Data Fig. 6 Distribution of cases with a first-degree family history of prostate cancer by GRS decile and population.

The percentage of family history-positive cases in each genetic risk score (GRS) category are shown in men of European and African ancestry. The x-axis indicates the GRS category and the y-axis is the percentage of family history-positive prostate cancer cases.

Extended Data Fig. 7 Comparison of the GRS distributions between cases and controls.

a) Men of European ancestry, b) Men of African Ancestry, c) Men of Asian ancestry and d) Hispanic men. The x-axis indicates the relative risk calculated by exponentiation of the difference in the mean genetic risk score (GRS) in controls and the mean GRS in cases for each population. The y-axis indicates the GRS density. Solid areas and corresponding percentages are the proportion of cases and controls with a GRS above 20% in the controls.

Extended Data Fig. 8 Distribution of aggressive and non-aggressive prostate cancer cases by GRS category.

a) Men of European ancestry and b) Men of African ancestry. The x-axis indicates the percentage of aggressive or non-aggressive prostate cancer cases and the y-axis indicates the genetic risk score (GRS) category.

Extended Data Fig. 9 Absolute risks of prostate cancer by GRS category.

a) Men of European ancestry from the UK Biobank and b) Men of African ancestry from the California Uganda (CA UG) study. The x-axis indicates the age of an individual and the y-axis indicates the absolute risk by a given age. Colored lines correspond to the indicated genetic risk score (GRS) category.

Extended Data Fig. 10 Absolute risks of prostate cancer by GRS category including individuals with a positive first-degree family history for prostate cancer (FH+).

a) Men of European ancestry and b) Men of African ancestry. The x-axis indicates the age of an individual and the y-axis indicates the absolute risk by a given age. Colored lines correspond to the indicated genetic risk score (GRS) category. FH+: family history positive.

Supplementary information

Supplementary Information

Supplementary Figs. 1 and 2

Reporting Summary

Supplementary Tables

Supplementary Tables 1–26

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Conti, D.V., Darst, B.F., Moss, L.C. et al. Trans-ancestry genome-wide association meta-analysis of prostate cancer identifies new susceptibility loci and informs genetic risk prediction. Nat Genet 53, 65–75 (2021). https://doi.org/10.1038/s41588-020-00748-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-020-00748-0

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing