Author Manuscript Published OnlineFirst on June 1, 2015; DOI: 10.1158/2159-8290.CD-15-0315 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

A large multi-ethnic genome-wide association study of prostate cancer identifies novel risk variants and substantial ethnic differences

Thomas J. Hoffmann,1,2 Stephen K. Van Den Eeden,3,4 Lori C. Sakoda,3 Eric Jorgenson,3 Laurel A. Habel,3 Rebecca E. Graff,1 Michael N. Passarelli,1 Clinton L. Cario,1 Nima C. Emami,1 Chun R. Chao,5 Nirupa R. Ghai,5 Jun Shan,3 Dilrini K. Ranatunga,3 Charles P. Quesenberry,3 David Aaronson,6 Joseph Presti,6 Wang Zhaoming,7 Sonja I. Berndt,7 Stephen J. Chanock,7 Shannon K. McDonnell,8 Amy J French,9 Daniel J Schaid,8 Stephen N. Thibodeau,9 Qiyuan Li,10 Matthew L. Freedman,11 Kathryn L. Penney,12 Lorelei A. Mucci,12 Christopher A. Haiman,13 Brian E. Henderson,13 Daniela Seminara,14 Mark N. Kvale,2 Pui-Yan Kwok,2 Catherine Schaefer,3 Neil Risch,1,2,3 and John S. Witte1,2,4,15

1

Department of Epidemiology and Biostatistics, University of California San Francisco, San

Francisco, CA 94158, USA 2

Institute for Human Genetics, University of California San Francisco, San Francisco, CA, 94143

USA 3

Division of Research, Kaiser Permanente, Northern California, Oakland, CA 94612, USA

4

Department of Urology, University of California San Francisco, San Francisco, CA 94158, USA

5

Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena CA

91101, USA 6

Department of Urology, Kaiser Oakland Medical Center, Northern California, Oakland, CA

94612, USA

1

Downloaded from cancerdiscovery.aacrjournals.org on June 6, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 1, 2015; DOI: 10.1158/2159-8290.CD-15-0315 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

7

Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics,

Department of Health and Human Services, National Cancer Institute, National Institutes of Health, Bethesda MD, USA. 8Department

of Health Science Research, Mayo Clinic, Rochester MN

9Department

of Laboratory Medicine and Pathology, Mayo Clinic, Rochester MN

10Medical

College, Xiamen University, Xiamen China 361102

11Department

of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical

School, Boston, MA 02115 and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA and The Eli and Edythe L. Broad Institute, Cambridge, MA 12Department

of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA and

Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 13Department

of Preventive Medicine, Keck School of Medicine and Norris Comprehensive

Cancer Center, University of Southern California, Los Angeles, CA 14National 15

Cancer Institute, National Institutes of Health, Bethesda, MD

UCSF Helen Diller Family Comprehensive Cancer Center, University of California San

Francisco, San Francisco, CA 94158, USA

Corresponding Authors: John S. Witte, University of California San Francisco, 1450 3rd St, San Francisco, CA 94158, USA, Tel: 415-502-6882, Fax: 415-476-1356 ([email protected]) or Stephen Van Den Eeden, Division of Research, Kaiser Permanente, Northern California, Oakland, CA 94612, Tel: 510-891-3718

2

Downloaded from cancerdiscovery.aacrjournals.org on June 6, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 1, 2015; DOI: 10.1158/2159-8290.CD-15-0315 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

([email protected])

Running title: GWAS of Prostate Cancer Keywords: Prostate cancer, genome-wide association study Abbreviations list: PCa, prostate cancer; KP, Kaiser Permanente; RPGEH, Research Program on Genes, Environment, and Health; MEC, Multiethnic Cohort; PSA, prostate specific antigen; LD, linkage disequilibrium; BMI, body mass index; DHSs, DNaseI hypersensitivity sites; CGEMs, Cancer Genemics Markers of Susceptibility Project; CMHS, California Men's Health Study; GERA, Genetic Epidemiology Research on Aging; KPNCCR, Kaiser Permanente Northern California Cancer Registry; KPSCCR, Kaiser Permanente Southern California Cancer Registry; SEER, Surveillance, Epidemiology, and End Results; EUR, European-optimized array; EAS, East Asian-optimized array; LAT, Latino-optimized array; AFR, African-Americanoptimized array; QC, quality control; MAF, minor allele frequency; HWE, Hardy-Weinberg Equilibrium; PCs, Principal components; H&E, hemetoxylin and eosin; PHS, Physician's Health Study; HPFS, Healthy Professionals Follow-up study; BPC3, Breast and Prostate Cancer Cohort Consortium; TCGA, The Cancer Genome Atlas; FE, fixed effects; RE, random effects; CI, confidence interval

Notes: Grant support: This work was supported by National Institutes of Health grants: CA127298, CA088164 and CA112355 (JSW); AG046616 and DC013300 (TJH); R01 GM107427 (MLF); CA136578, CA141298, and CA131945 (KLP, LAM); CA151254 and CA157881 (SNT). This work was also 3

Downloaded from cancerdiscovery.aacrjournals.org on June 6, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 1, 2015; DOI: 10.1158/2159-8290.CD-15-0315 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

supported by the UCSF Goldberg-Benioff Program in Cancer Translational Biology (JSW), Prostate Cancer Foundation Young Investigator Awards (KLP, LAM), Department Of Defense grant W81XWH-11-1-0261 (SNT), the MEC and the genotyping in AAPC were supported by NIH grants CA63464, CA54281, CA1326792, CA148085 and HG004726, funds from the California Cancer Research Program, grant number 99-86883, and from The Community Benefit Program, Kaiser Permanente Northern California. Support for participant enrollment, survey completion, and biospecimen collection for the RPGEH was provided by the Robert Wood Johnson Foundation, the Wayne and Gladys Valley Foundation, the Ellison Medical Foundation, and Kaiser Permanente national and regional community benefit programs. Genotyping of the GERA cohort was funded by a grant from the National Institute on Aging, National Institute of Mental Health, and the National Institute of Health Common Fund (RC2 AG036607 to CAS and NJR). PEGASUS was supported by the Intramural Research Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. Reprints requests and fees payment: John S. Witte, University of California San Francisco, 1450 3rd St, San Francisco, CA 94158, USA, Tel: 415-502-6882, Fax: 415-476-1356 ([email protected]) Conflict of Interest: No potential conflicts of interest were disclosed.

4

Downloaded from cancerdiscovery.aacrjournals.org on June 6, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 1, 2015; DOI: 10.1158/2159-8290.CD-15-0315 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Abstract A genome-wide association study of prostate cancer in Kaiser Permanente health plan members (7,783 cases, 38,595 controls; 80.3% non-Hispanic white, 4.9% African-American, 7.0% East Asian, 7.8% Latino) revealed a new independent risk indel rs4646284 at the previouslyidentified locus 6q25.3 that replicated in PEGASUS (N=7,539) and MEC (N=4,679) (p=1.0x1019

, OR=1.18). Across the 6q25.3 locus, rs4646284 exhibited the strongest association with

expression of SLC22A1 (p=1.3x10-23) and SLC22A3 (p=3.2x10-52). At the known 19q13.33 locus rs2659124 (p=1.3x10-13, OR=1.18) nominally replicated in PEGASUS. A risk score of 105 known risk SNPs was strongly associated with prostate cancer (p1.0). Only four of the variants with ORs0.3, so all were used. In the East Asian race/ethnicity group, rs116041037 and rs7210100 had r2=0.87, and rs1016343 and rs6983562 has r2=0.55; the latter SNP was removed in both circumstances when computing the risk score. To estimate the variance explained by these risk scores, we report Nagelgerke's pseudo-r2 estimate (57).

GWAS Array Heritability We estimated the additive array heritability using GCTA v1.24 (58). Array heritability estimates can be more sensitive to artifacts than GWAS results (58). Thus, to limit any potential batch effects, we limited this analysis to the homogeneous group of 30,598 non-Hispanic white men (3,605 cases and 26,993 controls) genotyped with the EUR array with Axiom v1 reagent kit. We also employed a stronger set of filters than used in the GWAS (58). Specifically, in addition to the filters noted above, we excluded SNPs with: HWE p0.05. We used only the autosomes, as is commonly done for estimating heritability. Finally, we LD-filtered the SNPs such that no two SNPs had a pairwise r2>0.8. This resulted in 26,226 individuals (3,143 cases and 23,083 controls), 402,748 genotyped SNPs, and 2,184,083 imputed SNPs with rinfo2≥0.3 and

28

Downloaded from cancerdiscovery.aacrjournals.org on June 6, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 1, 2015; DOI: 10.1158/2159-8290.CD-15-0315 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

MAF≥0.01 used in the analysis. We assumed a population prevalence of PCa=0.12 in the liability threshold model.

We further partitioned the genome into several sets using a joint variance components fit in GCTA (59). We first tested the previously-known hits vs. the rest of the genome, and then partitioned into the functional categories, prioritized similarly to (33): coding, UTR, promoter, DHS, intron, and intergenic. SNPs that happened to fall in overlapping regions were assigned to the highest priority category. The coding, UTR, and intron regions were determined from the UCSC Genome Browser known gene database (60). Unlike (33), who defined the promoters as +/-2Kb of a transcription start site, we defined the promoters from the Eukaryotic Promoter Database new v003 (34). The DHSs were determined from (33). For this partitioning, we defined enrichment for each category as the percentage of heritability explained divided by the percentage of genome. CIs and p-values were determined by 108 bootstrap iterations.

29

Downloaded from cancerdiscovery.aacrjournals.org on June 6, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 1, 2015; DOI: 10.1158/2159-8290.CD-15-0315 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Acknowledgements We are grateful to the Kaiser Permanente Northern California members who have generously agreed to participate in the Kaiser Permanente Research Program on Genes, Environment, and Health, the ProHealth Study and the California Men's Health Study. We would like to thank Peter Kraft, David Hunter, Sara Lindstrom, and Constance Chen for the contribution of PHS and HPFS genetic data through the BPC3.

30

Downloaded from cancerdiscovery.aacrjournals.org on June 6, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 1, 2015; DOI: 10.1158/2159-8290.CD-15-0315 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

References

1.

Hayes RB, Liff JM, Pottern LM, Greenberg RS, Schoenberg JB, Schwartz AG, et al. Prostate cancer risk in U.S. blacks and whites with a family history of cancer. Int J Cancer J Int Cancer. 1995;60:361–4.

2.

Whittemore AS, Wu AH, Kolonel LN, John EM, Gallagher RP, Howe GR, et al. Family history and prostate cancer risk in black, white, and Asian men in the United States and Canada. Am J Epidemiol. 1995;141:732–40.

3.

Schaid DJ. The complex genetic epidemiology of prostate cancer. Hum Mol Genet. 2004;13 Spec No 1:R103–21.

4.

Baker SG, Lichtenstein P, Kaprio J, Holm N. Genetic susceptibility to prostate, breast, and colorectal cancer among Nordic twins. Biometrics. 2005;61:55–63.

5.

Hjelmborg JB, Scheike T, Holst K, Skytthe A, Penney KL, Graff RE, et al. The heritability of prostate cancer in the nordic twin study of cancer. Cancer Epidemiol Biomark Prev Publ Am Assoc Cancer Res Cosponsored Am Soc Prev Oncol. 2014;23:2303–10.

6.

Lichtenstein P, Holm NV, Verkasalo PK, Iliadou A, Kaprio J, Koskenvuo M, et al. Environmental and Heritable Factors in the Causation of Cancer — Analyses of Cohorts of Twins from Sweden, Denmark, and Finland. N Engl J Med. 2000;343:78–85.

7.

Akamatsu S, Takata R, Haiman CA, Takahashi A, Inoue T, Kubo M, et al. Common variants at 11q12, 10q26 and 3p11.2 are associated with prostate cancer susceptibility in 31

Downloaded from cancerdiscovery.aacrjournals.org on June 6, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 1, 2015; DOI: 10.1158/2159-8290.CD-15-0315 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Japanese. Nat Genet. 2012;44:426–9, S1. 8.

Cheng I, Chen GK, Nakagawa H, He J, Wan P, Laurie CC, et al. Evaluating Genetic Risk for Prostate Cancer among Japanese and Latinos. Cancer Epidemiol Biomarkers Prev. 2012;21:2048–58.

9.

Eeles RA, Kote-Jarai Z, Olama AAA, Giles GG, Guy M, Severi G, et al. Identification of seven new prostate cancer susceptibility loci through a genome-wide association study. Nat Genet. 2009;41:1116–21.

10. Eeles RA, Olama AAA, Benlloch S, Saunders EJ, Leongamornlert DA, Tymrakiewicz M, et al. Identification of 23 new prostate cancer susceptibility loci using the iCOGS custom genotyping array. Nat Genet. 2013;45:385-91. 11. Gudmundsson J, Sulem P, Manolescu A, Amundadottir LT, Gudbjartsson D, Helgason A, et al. Genome-wide association study identifies a second prostate cancer susceptibility variant at 8q24. Nat Genet. 2007;39:631–7. 12. Gudmundsson J, Sulem P, Rafnar T, Bergthorsson JT, Manolescu A, Gudbjartsson D, et al. Common sequence variants on 2p15 and Xp11.22 confer susceptibility to prostate cancer. Nat Genet. 2008;40:281–3. 13. Gudmundsson J, Sulem P, Gudbjartsson DF, Blondal T, Gylfason A, Agnarsson BA, et al. Genome-wide association and replication studies identify four variants associated with prostate cancer susceptibility. Nat Genet. 2009;41:1122–6.

32

Downloaded from cancerdiscovery.aacrjournals.org on June 6, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 1, 2015; DOI: 10.1158/2159-8290.CD-15-0315 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

14. Haiman CA, Patterson N, Freedman ML, Myers SR, Pike MC, Waliszewska A, et al. Multiple regions within 8q24 independently affect risk for prostate cancer. Nat Genet. 2007;39:638–44. 15. Haiman CA, Chen GK, Blot WJ, Strom SS, Berndt SI, Kittles RA, et al. Genome-wide association study of prostate cancer in men of African ancestry identifies a susceptibility locus at 17q21. Nat Genet. 2011;43:570–3. 16. Han Y, Signorello LB, Strom SS, Kittles RA, Rybicki BA, Stanford JL, et al. Generalizability of established prostate cancer risk variants in men of African ancestry. Int J Cancer. 2015;136:1210–7. 17. Jia L, Landan G, Pomerantz M, Jaschek R, Herman P, Reich D, et al. Functional Enhancers at the Gene-Poor 8q24 Cancer-Linked Locus. PLoS Genet. 2009;5:e1000597. 18. Kote-Jarai Z, Olama AAA, Giles GG, Severi G, Schleutker J, Weischer M, et al. Seven novel prostate cancer susceptibility loci identified by a multi-stage genome-wide association study. Nat Genet. 2011;43:785–91. 19. Lindstrom S, Schumacher F, Siddiq A, Travis RC, Campa D, Berndt SI, et al. Characterizing Associations and SNP-Environment Interactions for GWAS-Identified Prostate Cancer Risk Markers—Results from BPC3. PLoS ONE. 2011;6:e17142. 20. Lindström S, Schumacher FR, Campa D, Albanes D, Andriole G, Berndt SI, et al. Replication of Five Prostate Cancer Loci Identified in an Asian Population—Results from the NCI Breast and Prostate Cancer Cohort Consortium (BPC3). Cancer Epidemiol 33

Downloaded from cancerdiscovery.aacrjournals.org on June 6, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 1, 2015; DOI: 10.1158/2159-8290.CD-15-0315 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Biomarkers Prev. 2012;21:212–6. 21. Olama AAA, Kote-Jarai Z, Schumacher FR, Wiklund F, Berndt SI, Benlloch S, et al. A meta-analysis of genome-wide association studies to identify prostate cancer susceptibility loci associated with aggressive and non-aggressive disease. Hum Mol Genet. 2013;22:408– 15. 22. Al Olama AA, Kote-Jarai Z, Giles GG, Guy M, Morrison J, Severi G, et al. Multiple loci on 8q24 associated with prostate cancer susceptibility. Nat Genet. 2009;41:1058–60. 23. Schumacher FR, Berndt SI, Siddiq A, Jacobs KB, Wang Z, Lindstrom S, et al. Genomewide association study identifies new prostate cancer susceptibility loci. Hum Mol Genet. 2011;20:3867–75. 24. Sun J, Zheng SL, Wiklund F, Isaacs SD, Li G, Wiley KE, et al. Sequence Variants at 22q13 Are Associated with Prostate Cancer Risk. Cancer Res. 2009;69:10–5. 25. Takata R, Akamatsu S, Kubo M, Takahashi A, Hosono N, Kawaguchi T, et al. Genomewide association study identifies five new susceptibility loci for prostate cancer in the Japanese population. Nat Genet. 2010;42:751–4. 26. Thomas G, Jacobs KB, Yeager M, Kraft P, Wacholder S, Orr N, et al. Multiple loci identified in a genome-wide association study of prostate cancer. Nat Genet. 2008;40:310– 5. 27. Xu J, Mo Z, Ye D, Wang M, Liu F, Jin G, et al. Genome-wide association study in Chinese

34

Downloaded from cancerdiscovery.aacrjournals.org on June 6, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 1, 2015; DOI: 10.1158/2159-8290.CD-15-0315 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

men identifies two new prostate cancer risk loci at 9q31.2 and 19q13.4. Nat Genet. 2012;44:1231–5. 28. Yeager M, Orr N, Hayes RB, Jacobs KB, Kraft P, Wacholder S, et al. Genome-wide association study of prostate cancer identifies a second risk locus at 8q24. Nat Genet. 2007;39:645–9. 29. Enger SM, Van Den Eeden SK, Sternfeld B, Loo RK, Quesenberry CP, Rowell S, et al. California Men’s Health Study (CMHS): a multiethnic cohort in a managed care setting. BMC Public Health. 2006;6:172. 30. Hoffmann TJ, Zhan Y, Kvale MN, Hesselson SE, Gollub J, Iribarren C, et al. Design and coverage of high throughput genotyping arrays optimized for individuals of East Asian, African American, and Latino race/ethnicity using imputation and a novel hybrid SNP selection algorithm. Genomics. 2011;98:422–30. 31. Hoffmann TJ, Kvale MN, Hesselson SE, Zhan Y, Aquino C, Cao Y, et al. Next generation genome-wide association tool: Design and coverage of a high-throughput Europeanoptimized SNP array. Genomics. 2011;98:79–89. 32. Byrne CD, Schwartz K, Lawn RM. Loss of a splice donor site at a “skipped exon” in a gene homologous to apolipoprotein(a) leads to an mRNA encoding a protein consisting of a single kringle domain. Arterioscler Thromb Vasc Biol. 1995;15:65–70. 33. Gusev A, Lee SH, Trynka G, Finucane H, Vilhjálmsson BJ, Xu H, et al. Partitioning Heritability of Regulatory and Cell-Type-Specific Variants across 11 Common Diseases. 35

Downloaded from cancerdiscovery.aacrjournals.org on June 6, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 1, 2015; DOI: 10.1158/2159-8290.CD-15-0315 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Am J Hum Genet. 2014;95:535–52. 34. Dreos R, Ambrosini G, Périer RC, Bucher P. The Eukaryotic Promoter Database: expansion of EPDnew and new promoter analysis tools. Nucleic Acids Res. 2015;43:D92–6. 35. Parikh H, Wang Z, Pettigrew KA, Jia J, Daugherty S, Yeager M, et al. Fine mapping the KLK3 locus on chromosome 19q13.33 associated with prostate cancer susceptibility and PSA levels. Hum Genet. 2011;129:675–85. 36. Sun J, Tao S, Gao Y, Peng T, Tan A, Zhang H, et al. Genome-wide association study identified novel genetic variant on SLC45A3 gene associated with serum levels prostatespecific antigen (PSA) in a Chinese population. Hum Genet. 2013;132:423–9. 37. Ahn J, Berndt SI, Wacholder S, Kraft P, Kibel AS, Yeager M, et al. Variation in KLK Genes, Prostate Specific Antigen, and Risk of Prostate Cancer. Nat Genet. 2008;40:1032-4. 38. He Y, Gu J, Strom S, Logothetis CJ, Kim J, Wu X. The prostate cancer susceptibility variant rs2735839 near KLK3 gene is associated with aggressive prostate cancer and can stratify gleason score 7 patients. Clin Cancer Res Off J Am Assoc Cancer Res. 2014;20:5133–9. 39. Kraft P. Curses—Winnerʼs and Otherwise—in Genetic Epidemiology. Epidemiology. 2008;19:649–51. 40. Zaitlen N, Kraft P, Patterson N, Pasaniuc B, Bhatia G, Pollack S, et al. Using Extended Genealogy to Estimate Components of Heritability for 23 Quantitative and Dichotomous

36

Downloaded from cancerdiscovery.aacrjournals.org on June 6, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 1, 2015; DOI: 10.1158/2159-8290.CD-15-0315 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Traits. PLoS Genet. 2013;9:e1003520. 41. Hoffmann TJ, Sakoda LC, Shen L, Jorgenson E, Habel LA, Liu J, et al. Imputation of the Rare HOXB13 G84E Mutation and Cancer Risk in a Large Population-Based Cohort. PLoS Genet. 2015;11:e1004930. 42. Kolonel LN, Henderson BE, Hankin JH, Nomura AM, Wilkens LR, Pike MC, et al. A multiethnic cohort in Hawaii and Los Angeles: baseline characteristics. Am J Epidemiol. 2000;151:346–57. 43. Delaneau O, Marchini J, Zagury J-F. A linear complexity phasing method for thousands of genomes. Nat Methods. 2012;9:179–81. 44. Howie B, Marchini J, Stephens M. Genotype Imputation with Thousands of Genomes. G3 Genes Genomes Genet. 2011;1:457–70. 45. Howie B, Fuchsberger C, Stephens M, Marchini J, Abecasis GR. Fast and accurate genotype imputation in genome-wide association studies through pre-phasing. Nat Genet. 2012;44:955–9. 46. Howie BN, Donnelly P, Marchini J. A Flexible and Accurate Genotype Imputation Method for the Next Generation of Genome-Wide Association Studies. PLoS Genet. 2009;5:e1000529. 47. Marchini J, Howie B. Genotype imputation for genome-wide association studies. Nat Rev Genet. 2010;11:499–511.

37

Downloaded from cancerdiscovery.aacrjournals.org on June 6, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 1, 2015; DOI: 10.1158/2159-8290.CD-15-0315 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

48. Zheng J, Li Y, Abecasis GR, Scheet P. A comparison of approaches to account for uncertainty in analysis of imputed genotypes. Genet Epidemiol. 2011;35:102–10. 49. Price AL, Patterson NJ, Plenge RM, Weinblatt ME, Shadick NA, Reich D. Principal components analysis corrects for stratification in genome-wide association studies. Nat Genet. 2006;38:904–9. 50. Devlin B, Roeder K. Genomic control for association studies. Biometrics. 2004;55:997– 1004. 51. Han B, Eskin E. Random-Effects Model Aimed at Discovering Associations in MetaAnalysis of Genome-wide Association Studies. Am J Hum Genet. 2011;88:586–98. 52. Larson NB, McDonnel S, French AJ, Fogarty Z, Cheville J, Middha S, et al. Am J Hum Genet. 2015; 10.1016/j.ajhg.2015.04.015. 53. Penney KL, Sinnott JA, Tyekucheva S, Gerke T, Shui IM, Kraft P, et al. Association of prostate cancer risk variants with gene expression in normal and tumor tissue. Cancer Epidemiol Biomark Prev Publ Am Assoc Cancer Res Cosponsored Am Soc Prev Oncol. 2015;24:255–60. 54. Taylor BS, Schultz N, Hieronymus H, Gopalan A, Xiao Y, Carver BS, et al. Integrative genomic profiling of human prostate cancer. Cancer Cell. 2010;18:11–22. 55. Li Q, Stram A, Chen C, Kar S, Gayther S, Pharoah P, et al. Expression QTL-based analyses reveal candidate causal genes and loci across five tumor types. Hum Mol Genet.

38

Downloaded from cancerdiscovery.aacrjournals.org on June 6, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 1, 2015; DOI: 10.1158/2159-8290.CD-15-0315 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

2014;23:5294–302. 56. Qiu W, Lazarus R. GeneticsDesign: Functions for designing genetic studies. R Package version 1.28.0. 2010. 57. Nagelkerke NJD. A note on a general definition of the coefficient of determination. Biometrika. 1991;78:691–2. 58. Lee SH, Wray NR, Goddard ME, Visscher PM. Estimating Missing Heritability for Disease from Genome-wide Association Studies. Am J Hum Genet. 2011;88:294–305. 59. Yang J, Lee SH, Goddard ME, Visscher PM. GCTA: A Tool for Genome-wide Complex Trait Analysis. Am J Hum Genet. 2011;88:76–82. 60. Karolchik D, Barber GP, Casper J, Clawson H, Cline MS, Diekhans M, et al. The UCSC Genome Browser database: 2014 update. Nucleic Acids Res. 2014;42:D764–70.

39

Downloaded from cancerdiscovery.aacrjournals.org on June 6, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 1, 2015; DOI: 10.1158/2159-8290.CD-15-0315 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Tables

Table 1. Descriptive factors for KP study population used in genome-wide association study of prostate cancer.

Cases N Total Race/ethnicity: non-Hispanic white African-American Asian Latino Age: Age < 50 50 ≤ Age < 60 60 ≤Age < 70 70 ≤ Age PSA* Mean (SD) Median (MAD) Interquartile Range Gleason: ≤5 6 7 8 9 10

%

7783

Controls N

%

38595

6406 601 288 488

82.3% 7.7% 3.7% 6.3%

30866 1650 2938 3141

80.0% 4.3% 7.6% 8.1%

86 1224 3604 2869

1.1% 15.7% 46.3% 36.9%

4619 7536 12240 14200

12.0% 19.5% 31.7% 36.8%

11.6 6.4 4.8-9.7

(53.4) (3.0)

2.5 1.5 0.8-2.8

(5.5) (1.2)

167 3067 1641 292 174 27

2.3% 53.8% 33.1% 5.8% 3.5% 0.5%

-------------

-------------

* PSA is given as the latest measure before diagnosis for cases, and the latest measure available to us from electronic medical records for controls. MAD, median absolute deviation.

40

Downloaded from cancerdiscovery.aacrjournals.org on June 6, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 1, 2015; DOI: 10.1158/2159-8290.CD-15-0315 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Table 2. Prostate cancer genome-wide association study results for new risk indel rs4646284 at 6q25.3 and for risk SNP rs2659124 at 19q13.33. These variants were detected in the KP study population and tested for replication in the PEGASUS and MEC cohorts. (a) Single variant and conditional analyses for 6q25.3 indel rs4646284 and the previous prostate cancer risk SNPs rs9364554 and rs651164. (b) Single variant and conditional analyses for 19q13.33 SNP rs2659124 detected here and the previous prostate cancer risk SNP rs2735839. Meta-analysis is given for the fixed effects (FE) and random effects (RE) analysis. Chromosome positions are given in b37. The risk allele frequency (RAF) is the pooled frequency across both cases and controls.

(a) No Conditioning Variant

OR (95% CI)

p

OR (95% CI)

p

r2info

KP NH white KP Latino KP Asian KP Af-Amer KP Meta FE KP Meta RE MEC PEGASUS Overall Meta FE Overall Meta RE

1.17 (1.12, 1.23) 1.13 (0.94, 1.36) 1.03 (0.84, 1.27) 1.18 (1.02, 1.37) 1.17 (1.12, 1.22) 1.17 (1.12, 1.22) 1.13 (1.03, 1.25) 1.27 (1.17, 1.37) 1.18 (1.14, 1.22) 1.18 (1.13, 1.23)

6.7x10-11 0.20 0.76 0.029 5.5x10-12 5.5x10-12 0.0094 1.4x10-8 1.0x10-19 9.8x10-17

1.16 (1.10, 1.23) 1.06 (0.85, 1.31) 0.91 (0.68, 1.23) 1.21 (1.03, 1.42) 1.15 (1.09, 1.21) 1.15 (1.07, 1.22) 1.13 (1.03, 1.25) 1.20 (1.09, 1.32) 1.16 (1.11, 1.21) 1.16 (1.11, 1.21)

3.3x10-7 0.61 0.56 0.020 8.5x10-8 7.7x10-5 0.014 0.00024 5.4x10-12 5.4x10-12

0.91 0.89 0.89 0.85 0.81 0.80 -

C

KP NH white KP Latino KP Asian KP Af-Amer KP Meta FE KP Meta RE MEC PEGASUS Overall Meta FE Overall Meta RE

0.279 0.227 0.319 0.079 0.074 0.293 -

1.10 (1.05, 1.15) 1.16 (0.97, 1.29) 1.06 (0.87, 1.29) 1.14 (0.89, 1.47) 1.10 (1.06, 1.15) 1.10 (1.06, 1.15) 1.21 (1.02, 1.41) 1.22 (1.13, 1.31) 1.14 (1.10, 1.18) 1.15 (1.09, 1.20)

6.6x10-5 0.1 0.59 0.3 1.1x10-5 1.1x10-5 0.024 9.3x10-8 6.3x10-12 6.1x10-8

1.05 (1.00, 1.10) 1.13 (0.93, 1.36) 1.05 (0.86, 1.28) 1.11 (0.85, 1.43) 1.06 (1.01, 1.11) 1.06 (1.01, 1.11) 1.19 (1.00, 1.40) 1.16 (1.07, 1.25) 1.09 (1.05, 1.13) 1.10 (1.05, 1.15)

0.070 0.21 0.63 0.45 0.019 0.019 0.041 0.00022 1.9x10-5 0.00014

1.00 1.00 1.00 1.00 1.00 1.00 -

G

KP NH white KP Latino KP Asian KP Af-Amer KP Meta FE KP Meta RE MEC PEGASUS

0.308 0.380 0.515 0.264 0.307 0.306

0.94 (0.90, 0.98) 0.88 (0.75, 1.04) 0.91 (0.76, 1.10) 1.02 (0.88, 1.18) 0.94 (0.90, 0.98) 0.94 (0.90, 0.98) 0.98 (0.90, 1.08) 0.91 (0.84, 0.97)

0.0088 0.12 0.35 0.82 0.0037 0.0037 0.73 0.0065

1.01 (0.96, 1.06) 0.91 (0.76, 1.09) 0.85 (0.65, 1.11) 1.08 (0.93, 1.28) 1.00 (0.96, 1.05) 1.00 (0.93, 1.07) 1.03 (0.93, 1.13) 0.99 (0.92, 1.07)

0.79 0.29 0.24 0.30 0.96 0.90 0.60 0.82

1.00 1.00 1.00 1.00 1.00 1.00

Ref. Allele T

Previous SNP: rs9364554

T

Previous SNP: rs651164

A

Indel: rs4646284

Conditioning on other 6q25.3 SNPs

Risk AF 0.298 0.250 0.324 0.365 0.361 0.278 -

Risk Allele TG (indel)

Group

41

Downloaded from cancerdiscovery.aacrjournals.org on June 6, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 1, 2015; DOI: 10.1158/2159-8290.CD-15-0315 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Overall Meta FE Overall Meta RE

-

0.94 (0.91, 0.97) 0.94 (0.91, 0.97)

0.00019 0.00019

1.00 (0.97, 1.04) 1.00 (0.97, 1.04)

0.89 0.89

-

Pairwise correlations of these three SNPs in European, Latino, East Asian and AfricanAmericans, respectively (KP data): r2rs4646284,rs9364554

= 0.15, 0.090, 0.00028, 0.017;

r2rs4646284,rs651164

= 0.19, 0.19, 0.53, 0.10;

r2rs4646284,rs9364559

= 0.00047, 0.0088, 0.0018, 0.0010.

42

Downloaded from cancerdiscovery.aacrjournals.org on June 6, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 1, 2015; DOI: 10.1158/2159-8290.CD-15-0315 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

(b) No Conditioning SNP Risk SNP: rs2659124

Previous SNP: rs2735839

Risk Allele T

Ref. Allele A

T

C

Conditioning on other 19q13.33 SNPs

OR (95% CI)

p-value

OR (95% CI)

p-value

r2info

KP NH white KP Latino KP Asian KP Af-Amer KP Meta FE KP Meta RE MEC PEGASUS Overall Meta FE Overall Meta RE

Risk AF 0.854 0.809 0.587 0.847 0.839 0.868 -

1.22 (1.14, 1.30) 1.31 (1.07, 1.61) 1.15 (0.95, 1.40) 1.28 (1.05, 1.57) 1.22 (1.15, 1.29) 1.22 (1.15, 1.29) 1.06 (0.94, 1.20) 1.16 (1.05, 1.28) 1.18 (1.13, 1.24) 1.18 (1.12, 1.24)

2.2x10-9 0.0099 0.15 0.013 1.9x10-12 1.9x10-12 0.33 0.0027 1.3x10-13 3.0x10-10

1.13 (1.04, 1.23) 1.36 (1.00, 1.86) 1.03 (0.62, 1.71) 1.29 (1.05, 1.59) 1.16 (1.07, 1.25) 1.16 (1.07, 1.25) 1.09 (0.97, 1.23) 1.15 (1.01, 1.31) 1.14 (1.08, 1.21) 1.14 (1.08, 1.21)

0.0043 0.053 0.91 0.014 9.2x10-5 9.2x10-5 0.16 0.031 4.3x10-6 4.3x10-6

0.99 0.99 0.98 0.95 0.89 1.00 -

KP NH white KP Latino KP Asian KP Af-Amer KP Meta FE KP Meta RE MEC PEGASUS Overall Meta FE Overall Meta RE

0.150 0.214 0.419 0.302 0.311 0.140 -

1.20 (1.13, 1.28) 1.21 (1.00, 1.47) 1.15 (0.95, 1.40) 1.02 (0.88, 1.18) 1.17 (1.11, 1.24) 1.16 (1.07, 1.25) 0.93 (0.85, 1.02) 0.90 (0.82. 0.99) 0.88 (0.85, 0.92) 0.89 (0.84, 0.93)

1.6x10-8 0.052 0.15 0.70 5.7x10-9 0.00012 0.11 0.038 2.0x10-10 3.5x10-6

1.11 (1.02, 1.23) 0.97 (0.72, 1.30) 1.13 (0.68, 1.88) 0.98 (0.84, 1.14) 1.07 (1.00, 1.15) 1.07 (1.00, 1.15) 0.91 (0.83, 1.00) 0.99 (0.88, 1.13) 1.01 (0.96, 1.06) 1.00 (0.92, 1.09)

0.016 0.83 0.63 0.75 0.056 0.056 0.058 0.92 0.76 0.99

1.00 1.00 1.00 1.00 1.00 0.99 -

Study Group

Pairwise correlations of these two SNPs in European, Latino, East Asian and African-Americans, respectively (KP data): r2rs2659124, rs2735839 = 0.50, 0.58, 0.85, 0.055.

43

Downloaded from cancerdiscovery.aacrjournals.org on June 6, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 1, 2015; DOI: 10.1158/2159-8290.CD-15-0315 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Table 3. Array heritability enrichment of the coding, UTR, promoter, DHS, intron, and intergenic partitions for non-Hispanic whites using the imputed data. Results are from fitting each partition separately (univariate), and then subsequent results are from the joint multivariate fit, Kaiser Permanente Study Population.

Univariate

Multivariate fit

Region

hg2

hg2 (SE)

Coding

0.029 (0.014) 0.013 (0.014)

3.9%

18,608

0.85%

4.6 (0.0, 13.5)

0.45

UTR

0.048 (0.019) 0.016 (0.020)

4.7%

27,778

1.27%

3.7 (0.0, 12.2)

0.56

Promoter 0.001 (0.003) 0.001 (0.003)

0.2%

3.347

0.15%

1.3 (0.0, 10.8)

0.94

(SE)

% hg2

# SNPs

% Genome Enrichment (95% CI) Enrichment P

DHS

0.299 (0.049) 0.243 (0.065)

70.9%

361,426

16.56%

4.3 (2.0, 5.6)

0.0039

Intron

0.193 (0.045) 0.039 (0.055)

11.5%

699,518

32.04%

0.4 (0.0, 1.2)

0.14

8.7% 1,072,335

49.12%

0.2 (0.0, 0.8)

0.0058

0.334 (0.060) 0.342 (0.060) 100.0% 2,183,012

100.00%

1.0

Intergenic 0.199 (0.051) 0.030 (0.060) Total

-

44

Downloaded from cancerdiscovery.aacrjournals.org on June 6, 2015. © 2015 American Association for Cancer Research.

Author Manuscript Published OnlineFirst on June 1, 2015; DOI: 10.1158/2159-8290.CD-15-0315 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure Legends

Figure 1. Results from a genome-wide association study (GWAS) of prostate cancer in Kaiser Permanente population (8,399 cases and 38,745 controls), highlighting key chromosomal regions. P-values are for variant associations with prostate cancer from trans-ethnic fixed-effects meta-analysis of four race/ethnicity GWAS (non-Hispanic white, Latino, African-American, East Asian), each adjusted for age and ancestry principal components. Horizontal dashed lines indicated genome-wide statistical significance (p

A large multiethnic genome-wide association study of prostate cancer identifies novel risk variants and substantial ethnic differences.

A genome-wide association study (GWAS) of prostate cancer in Kaiser Permanente health plan members (7,783 cases, 38,595 controls; 80.3% non-Hispanic w...
3MB Sizes 0 Downloads 5 Views