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.

  • Letter
  • Published:

Identification of six new susceptibility loci for invasive epithelial ovarian cancer

Abstract

Genome-wide association studies (GWAS) have identified 12 epithelial ovarian cancer (EOC) susceptibility alleles. The pattern of association at these loci is consistent in BRCA1 and BRCA2 mutation carriers who are at high risk of EOC. After imputation to 1000 Genomes Project data, we assessed associations of 11 million genetic variants with EOC risk from 15,437 cases unselected for family history and 30,845 controls and from 15,252 BRCA1 mutation carriers and 8,211 BRCA2 mutation carriers (3,096 with ovarian cancer), and we combined the results in a meta-analysis. This new study design yielded increased statistical power, leading to the discovery of six new EOC susceptibility loci. Variants at 1p36 (nearest gene, WNT4), 4q26 (SYNPO2), 9q34.2 (ABO) and 17q11.2 (ATAD5) were associated with EOC risk, and at 1p34.3 (RSPO1) and 6p22.1 (GPX6) variants were specifically associated with the serous EOC subtype, all with P < 5 × 10−8. Incorporating these variants into risk assessment tools will improve clinical risk predictions for BRCA1 and BRCA2 mutation carriers.

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

Figure 1: HR estimates for association with EOC of 12 previously reported EOC susceptibility variants and the 6 new susceptibility variants for OCAC samples, BRCA1 mutation carriers and BRCA2 mutation carriers.
Figure 2: The 1p36 EOC susceptibility locus.

Similar content being viewed by others

References

  1. Auranen, A. et al. Cancer incidence in the first-degree relatives of ovarian cancer patients. Br. J. Cancer 74, 280–284 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Stratton, J.F., Pharoah, P., Smith, S.K., Easton, D. & Ponder, B.A. A systematic review and meta-analysis of family history and risk of ovarian cancer. Br. J. Obstet. Gynaecol. 105, 493–499 (1998).

    Article  CAS  PubMed  Google Scholar 

  3. Jervis, S. et al. Ovarian cancer familial relative risks by tumour subtypes and by known ovarian cancer genetic susceptibility variants. J. Med. Genet. 51, 108–113 (2014).

    Article  CAS  PubMed  Google Scholar 

  4. Bolton, K.L. et al. Common variants at 19p13 are associated with susceptibility to ovarian cancer. Nat. Genet. 42, 880–884 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Goode, E.L. et al. A genome-wide association study identifies susceptibility loci for ovarian cancer at 2q31 and 8q24. Nat. Genet. 42, 874–879 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Song, H. et al. A genome-wide association study identifies a new ovarian cancer susceptibility locus on 9p22.2. Nat. Genet. 41, 996–1000 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Pharoah, P.D. et al. GWAS meta-analysis and replication identifies three new susceptibility loci for ovarian cancer. Nat. Genet. 45, 362–370 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Permuth-Wey, J. et al. Identification and molecular characterization of a new ovarian cancer susceptibility locus at 17q21.31. Nat. Commun. 4, 1627 (2013).

    Article  CAS  PubMed  Google Scholar 

  9. Bojesen, S.E. et al. Multiple independent variants at the TERT locus are associated with telomere length and risks of breast and ovarian cancer. Nat. Genet. 45, 371–384 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Couch, F.J. et al. Common variants at the 19p13.1 and ZNF365 loci are associated with ER subtypes of breast cancer and ovarian cancer risk in BRCA1 and BRCA2 mutation carriers. Cancer Epidemiol. Biomarkers Prev. 21, 645–657 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Ramus, S.J. et al. Ovarian cancer susceptibility alleles and risk of ovarian cancer in BRCA1 and BRCA2 mutation carriers. Hum. Mutat. 33, 690–702 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Ramus, S.J. et al. Genetic variation at 9p22.2 and ovarian cancer risk for BRCA1 and BRCA2 mutation carriers. J. Natl. Cancer Inst. 103, 105–116 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Couch, F.J. et al. Genome-wide association study in BRCA1 mutation carriers identifies novel loci associated with breast and ovarian cancer risk. PLoS Genet. 9, e1003212 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Bolton, K.L., Ganda, C., Berchuck, A., Pharaoh, P.D. & Gayther, S.A. Role of common genetic variants in ovarian cancer susceptibility and outcome: progress to date from the Ovarian Cancer Association Consortium (OCAC). J. Intern. Med. 271, 366–378 (2012).

    Article  CAS  PubMed  Google Scholar 

  15. Chenevix-Trench, G. et al. An international initiative to identify genetic modifiers of cancer risk in BRCA1 and BRCA2 mutation carriers: the Consortium of Investigators of Modifiers of BRCA1 and BRCA2 (CIMBA). Breast Cancer Res. 9, 104 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Gaudet, M.M. et al. Identification of a BRCA2-specific modifier locus at 6p24 related to breast cancer risk. PLoS Genet. 9, e1003173 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. 1000 Genomes Project Consortium. An integrated map of genetic variation from 1,092 human genomes. Nature 491, 56–65 (2012).

  18. Barnes, D.R. et al. Evaluation of association methods for analysing modifiers of disease risk in carriers of high-risk mutations. Genet. Epidemiol. 36, 274–291 (2012).

    Article  PubMed  Google Scholar 

  19. Bourguignon, L.Y., Gilad, E., Rothman, K. & Peyrollier, K. Hyaluronan-CD44 interaction with IQGAP1 promotes Cdc42 and ERK signaling, leading to actin binding, Elk-1/estrogen receptor transcriptional activation, and ovarian cancer progression. J. Biol. Chem. 280, 11961–11972 (2005).

    Article  CAS  PubMed  Google Scholar 

  20. Zuo, Y., Wu, Y. & Chakraborty, C. Cdc42 negatively regulates intrinsic migration of highly aggressive breast cancer cells. J. Cell. Physiol. 227, 1399–1407 (2012).

    Article  CAS  PubMed  Google Scholar 

  21. Pagliardini, L. et al. An Italian association study and meta-analysis with previous GWAS confirm WNT4, CDKN2BAS and FN1 as the first identified susceptibility loci for endometriosis. J. Med. Genet. 50, 43–46 (2013).

    Article  CAS  PubMed  Google Scholar 

  22. Pearce, C.L. et al. Association between endometriosis and risk of histological subtypes of ovarian cancer: a pooled analysis of case-control studies. Lancet Oncol. 13, 385–394 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Tomaselli, S. et al. Human RSPO1/R-spondin1 is expressed during early ovary development and augments β-catenin signaling. PLoS ONE 6, e16366 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Parma, P. et al. R-spondin1 is essential in sex determination, skin differentiation and malignancy. Nat. Genet. 38, 1304–1309 (2006).

    Article  CAS  PubMed  Google Scholar 

  25. Tomizuka, K. et al. R-spondin1 plays an essential role in ovarian development through positively regulating Wnt-4 signaling. Hum. Mol. Genet. 17, 1278–1291 (2008).

    Article  CAS  PubMed  Google Scholar 

  26. De Ganck, A. et al. Multiple isoforms of the tumor suppressor myopodin are simultaneously transcribed in cancer cells. Biochem. Biophys. Res. Commun. 370, 269–273 (2008).

    Article  CAS  PubMed  Google Scholar 

  27. Jing, L. et al. Expression of myopodin induces suppression of tumor growth and metastasis. Am. J. Pathol. 164, 1799–1806 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Lin, F. et al. Myopodin, a synaptopodin homologue, is frequently deleted in invasive prostate cancers. Am. J. Pathol. 159, 1603–1612 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Sanchez-Carbayo, M., Schwarz, K., Charytonowicz, E., Cordon-Cardo, C. & Mundel, P. Tumor suppressor role for myopodin in bladder cancer: loss of nuclear expression of myopodin is cell-cycle dependent and predicts clinical outcome. Oncogene 22, 5298–5305 (2003).

    Article  CAS  PubMed  Google Scholar 

  30. Yu, Y.P. & Luo, J.H. Myopodin-mediated suppression of prostate cancer cell migration involves interaction with zyxin. Cancer Res. 66, 7414–7419 (2006).

    Article  CAS  PubMed  Google Scholar 

  31. Amundadottir, L. et al. Genome-wide association study identifies variants in the ABO locus associated with susceptibility to pancreatic cancer. Nat. Genet. 41, 986–990 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Rummel, S., Shriver, C.D. & Ellsworth, R.E. Relationships between the ABO blood group SNP rs505922 and breast cancer phenotypes: a genotype-phenotype correlation study. BMC Med. Genet. 13, 41 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Poole, E.M. et al. ABO blood group and risk of epithelial ovarian cancer within the Ovarian Cancer Association Consortium. Cancer Causes Control 23, 1805–1810 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Sikdar, N. et al. DNA damage responses by human ELG1 in S phase are important to maintain genomic integrity. Cell Cycle 8, 3199–3207 (2009).

    Article  CAS  PubMed  Google Scholar 

  35. Bell, D.W. et al. Predisposition to cancer caused by genetic and functional defects of mammalian Atad5. PLoS Genet. 7, e1002245 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Lee, K.Y. et al. Human ELG1 regulates the level of ubiquitinated proliferating cell nuclear antigen (PCNA) through Its interactions with PCNA and USP1. J. Biol. Chem. 285, 10362–10369 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Wacholder, S., Han, S.S. & Weinberg, C.R. Inference from a multiplicative model of joint genetic effects for ovarian cancer risk. J. Natl. Cancer Inst. 103, 82–83 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Hnisz, D. et al. Super-enhancers in the control of cell identity and disease. Cell 155, 934–947 (2013).

    Article  CAS  PubMed  Google Scholar 

  39. Permuth-Wey, J. et al. LIN28B polymorphisms influence susceptibility to epithelial ovarian cancer. Cancer Res. 71, 3896–3903 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Delaneau, O., Marchini, J. & Zagury, J.F. A linear complexity phasing method for thousands of genomes. Nat. Methods 9, 179–181 (2012).

    Article  CAS  Google Scholar 

  41. 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).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Antoniou, A.C. et al. RAD51 135G→C modifies breast cancer risk among BRCA2 mutation carriers: results from a combined analysis of 19 studies. Am. J. Hum. Genet. 81, 1186–1200 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Antoniou, A.C. et al. A locus on 19p13 modifies risk of breast cancer in BRCA1 mutation carriers and is associated with hormone receptor-negative breast cancer in the general population. Nat. Genet. 42, 885–892 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. R: A Language and Environment for Statistical Computing v. 3.0.1 (R Foundation for Statistical Computing, 2013).

  45. Mavaddat, N. et al. Pathology of breast and ovarian cancers among BRCA1 and BRCA2 mutation carriers: results from the Consortium of Investigators of Modifiers of BRCA1/2 (CIMBA). Cancer Epidemiol. Biomarkers Prev. 21, 134–147 (2012).

    Article  CAS  PubMed  Google Scholar 

  46. Lakhani, S.R. et al. Pathology of ovarian cancers in BRCA1 and BRCA2 carriers. Clin. Cancer Res. 10, 2473–2481 (2004).

    Article  CAS  PubMed  Google Scholar 

  47. Rubin, S.C. et al. Clinical and pathological features of ovarian cancer in women with germ-line mutations of BRCA1. N. Engl. J. Med. 335, 1413–1416 (1996).

    Article  CAS  PubMed  Google Scholar 

  48. Maehle, L. et al. High risk for ovarian cancer in a prospective series is restricted to BRCA1/2 mutation carriers. Clin. Cancer Res. 14, 7569–7573 (2008).

    Article  CAS  PubMed  Google Scholar 

  49. Shaw, P.A. et al. Histopathologic features of genetically determined ovarian cancer. Int. J. Gynecol. Pathol. 21, 407–411 (2002).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Udler, M.S., Tyrer, J. & Easton, D.F. Evaluating the power to discriminate between highly correlated SNPs in genetic association studies. Genet. Epidemiol. 34, 463–468 (2010).

    Article  PubMed  Google Scholar 

  52. Reumers, J. et al. Joint annotation of coding and non-coding single nucleotide polymorphisms and mutations in the SNPeffect and PupaSuite databases. Nucleic Acids Res. 36, D825–D829 (2008).

    Article  CAS  PubMed  Google Scholar 

  53. Conde, L. et al. PupaSuite: finding functional single nucleotide polymorphisms for large-scale genotyping purposes. Nucleic Acids Res. 34, W621–W625 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Li, N.F. et al. A modified medium that significantly improves the growth of human normal ovarian surface epithelial (OSE) cells in vitro. Lab. Invest. 84, 923–931 (2004).

    Article  PubMed  Google Scholar 

  55. Fotheringham, S., Levanon, K. & Drapkin, R. Ex vivo culture of primary human fallopian tube epithelial cells. J. Vis. Exp. 51, 2728 (2011).

    Google Scholar 

  56. Li, Q. et al. Expressopn QTL–based analyses reveal candidate causal genes and loci across five tumor types. Hum. Mol. Genet. 23, 5294–5302 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Cancer Genome Atlas Research Network. Integrated genomic analyses of ovarian carcinoma. Nature 474, 609–615 (2011).

  58. Forbes, S.A. et al. The Catalogue of Somatic Mutations in Cancer (COSMIC). Curr. Protoc. Hum. Genet. Chapter 10, Unit 10.11 (2008).

  59. Gao, J. et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal. 6, pl1 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Cerami, E. et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2, 401–404 (2012).

    Article  PubMed  Google Scholar 

  61. Lawrenson, K. et al. Senescent fibroblasts promote neoplastic transformation of partially transformed ovarian epithelial cells in a three-dimensional model of early stage ovarian cancer. Neoplasia 12, 317–325 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank all the individuals who took part in this study and all the researchers, clinicians, and technical and administrative staff who made possible the many studies contributing to this work (a full list is provided in the Supplementary Note), including X.Q. Chen for iPLEX genotyping. The COGS project is funded through a European Commission Seventh Framework Programme grant (agreement number 223175-HEALTH-F2-2009-223175). CIMBA data management and data analysis were supported by Cancer Research UK grants C12292/A11174 and C1287/A10118. The Ovarian Cancer Association Consortium (OCAC) is supported by a grant from the Ovarian Cancer Research Fund thanks to donations by the family and friends of Kathryn Sladek Smith (PPD/RPCI.07). Scientific development and funding for this project were in part supported by the US National Cancer Institute GAME-ON Post-GWAS Initiative (U19-CA148112). This study made use of data generated by the Wellcome Trust Case Control Consortium. Funding for the project was provided by the Wellcome Trust under award 076113. The results published here are in part based on data generated by The Cancer Genome Atlas (TCGA) Pilot Project established by the US National Cancer Institute and US National Human Genome Research Institute (database of Genotypes and Phenotypes (dbGaP) accession phs000178.v8.p7). The cBio Portal is developed and maintained by the Computational Biology Center at the Memorial Sloan-Kettering Cancer Center. S. Healey is supported by a National Health and Medical Research Council of Australia Program Grant to G.C.-T. Details of the funding of individual investigators and studies are provided in the Supplementary Note. A full list of the investigators who contributed to the generation of the data is available on the CIMBA website (see URLs).

Author information

Authors and Affiliations

Authors

Consortia

Contributions

Writing group: K.B.K., A.C.A., G.C.-T., S.J.R., J. Beesley, P.P.P., S.G. Performed statistical analyses for CIMBA: K.B.K. Performed statistical analyses for OCAC: J.T. Performed the meta-analyses: K.B.K. CIMBA database management: L.M. and D.B. Supervised CIMBA statistical analyses, meta-analyses and CIMBA data management: A.C.A. Supervised OCAC statistical analyses: P.P.P. Initiated and coordinated CIMBA: G.C.-T. Coordinated OCAC: A. Berchuck and P.P.P. Conceived and coordinated the synthesis of the iCOGS array: D.F.E. Coordinated iCOGS genotyping: J.S., K. Offit, F.J.C. iCOGS genotyping, calling and quality control: J.M. Cunningham, J.D., P.S., D.F.E., K.B.K., J.T., P.P.P., A.C.A., G.C.-T. Programming support: A. Lee. Provided DNA samples and/or phenotypic data: S.J.R., J.T., A. Lee, H.C.S., K.L., S. Healey, J.M.L., T.J.S., Y.G.L., T.P., Y.B., Q.L., S.C., D.H., A. Miron, M. Southey, M.B.T., D.E.G., S.S.B., R.J., C.M.D., E.J.v.R., S.L.N., Y.C.D., T.V.O.H., L.J., A.-M.G., B.E., J.D., J. Benitez, A.O., M.J.G., I. Komenaka, J.N.W., P.G., P.P., L. Bernard, A.V., B.B., B.P., S. Manoukian, P.R., L.P., L.O., F.F., I. Konstantopoulou, J. Garber, D.F., J. Perkins, R.P., S.E., EMBRACE, A.K.G., R.K.S., A. Meindl, C.E., C.S., O.M.S., GEMO, F.D., S. Mazoyer, D.S.-L., K. Claes, K.D.L., J. Kirk, G.C.R., M. Piedmonte, D.M.O., M.d.l.H., T.C., K.A., H. Nevanlinna, J.M. Collée, M.A. Rookus, J.C.O., F.B.L.H., HEBON, E.O., O.D., I.B., J. Brunet, C.L., M.A.P., A. Jakubowska, J. Gronwald, J. Lubinski, G.S., R.B.B., M. Plante, J.S., P.S., M.M., S. Tognazzo, M.R.T., KConFab, V.S.P., X. Wang, N.L., C.I.S., N.K., J.V., C.A.A., G.P., A. Berger, C.F.S., M.-K.T., C.M.P., M.H.G., P.L.M., G.R., A.M.M., S. Tchatchou, I.L.A., G.G., A.E.T., U.B.J., T.A.K., M. Thomassen, A. Bojesen, J.Z., E.F., Y.L., M. Soller, A. Liljegren, B.A., Z.E., M.S.-A., O.I.O., R.L.N., T.R.R., K.L.N., S.M.D., K.H.L., B.Y.K., C.W., J. Lester, Australian Cancer Society, Australian Ovarian Cancer Study Group, A.H., A.B.E., M.W.B., P.A.F., D. Lambrechts, E.V.N., I.V., S. Lambrechts, E.D., J.A.D., K.G.W., M.A. Rossing, A.R., J.C.-C., S.W.-G., U.E., K.B.M., K. Odunsi, L.S., S. Lele, L.R.W., M.T.G., P.J.T., Y.B.S., I.B.R., M.D., P. Hillemanns, T.D., N.A., N.B., A. Leminen, L.M.P., R.B., F.M., J.L.K., R.P.E., R.B.N., A.d.B., F.H., I.S., P. Harter, K.M., S. Hosono, S.O., A. Jensen, S.K.K., E.H., H.N.H., M.A.N.A., S.-H.T., Y.-L.W., B.L.F., E.L.G., J.M. Cunningham, R.A.V., F.B., G.G.G., D. Liang, M.A.T.H., X. Wu, D.A.L., M.B., A. Berchuck, E.S.I., J.M.S., P.C., R.P.W., D.W.C., K.L.T., E.M.P., S.S.T., E.V.B., I.O., S.H.O., C.K., H.B.S., I.L.T., L. Bjorge, A.M.v.A., K.K.H.A., L.A.K., L.F.A.G.M., M.K., A.B.-W., L.E.K., L.S.C., N.D.L., C.C., H.Y., J. Lissowska, L.A.B., N.W., C.H., L.L., L.N., H.B., H.S., D.E., I.G.C., I.M., J. Paul, K. Carty, N.S., R.G., A.S.W., J.H.R., V.M., W.S., B.-T.J., W.Z., X.-O.S., Y.-T.G., B.R., H.A.R., J.R.M., S.A.N., A.N.M., A.C., H.-Y.L., J.P.-W., T.A.S., Y.-Y.T., Z.C., A.Z., H.A.-C., A.G.-M., U.M., P. Harrington, A.W.L., A.H.W., C.L.P., G.C., M.C.P., A.D.-M., A.T., I.K.R., J. Kupryjanczyk, M.F., H. Noushmehr, L.T., N.T., U.H., C.I., M. Tischkowitz, E.N.I., M.A.C., D.F.E., K. Offit, F.J.C., S.G., P.P.P., A.C.A., G.C.-T. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Georgia Chenevix-Trench.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

A full list of members appears in the Supplementary Note.

A full list of members appears in the Supplementary Note.

A full list of members appears in the Supplementary Note.

A full list of members appears in the Supplementary Note.

A full list of members appears in the Supplementary Note.

A full list of members appears in the Supplementary Note.

A full list of members appears in the Supplementary Note.

A full list of members appears in the Supplementary Note.

Integrated supplementary information

Supplementary Figure 1 Imputation accuracy distribution.

Histogram showing the distribution of imputation accuracy estimates r2 in the first genotype imputation on the 1000 Genomes Project data v3 for SNPs with MAF > 0.05 (a,c,e) and for SNPs with MAF ≤ 0.05 (b,d,f) in OCAC-iCOGS (a,b), BRCA1 mutation carriers (c,d) and BRCA2 mutation carriers (e,f).

Supplementary Figure 2 Imputation accuracy distribution.

Histogram showing the distribution of imputation accuracy estimates r2 in the first genotype imputation on the 1000 Genomes Project data v3 for SNPs with MAF > 0.05 (a,c,e) and for SNPs with MAF ≤ 0.05 (b,d,f) in the UK GWAS (a,b), the US GWAS (c,d) and the Mayo GWAS (e,f).

Supplementary Figure 3 Quantile-quantile plot for genetic variants from the genotype imputation.

The column on the left shows all variants, and the right column shows variants not located in regions previously known to be associated with invasive ovarian cancer.

Supplementary Figure 4 Meta-analysis risk associations.

Manhattan plots showing the meta-analysis associations of genetic variants with risk of any subtype of ovarian cancer (a,b) and serous subtype ovarian cancer (c,d) for all genetic variants available after the first imputation (a,c) and after excluding SNPs located within known ovarian cancer susceptibility loci (b,d).

Supplementary Figure 5 Regional association plots for each novel locus based on the meta-analysis.

For 17q11.2, the meta-analysis was based on OCAC and BRCA2 mutation carriers only. For 1p34.3 and 6p22.1, the OCAC analysis was based on serous ovarian cancer. SNPs genotyped by the iCOGS array are shown in magenta, and imputed SNPs are shown in black.

Supplementary Figure 6 Ovarian cancer susceptibility loci at chromosome 1 and chromosome 4.

The Manhattan plot depicts the strength of association between all imputed and genotyped SNPs across the regions at chromosome 1 (a) and chromosome 4 (b). The dotted line represents the genome-wide significance level 5 × 10−8. FAIRE-seq data revealing potential regulatory regions in ovarian and fallopian tube cells are depicted as black bars. Additional tracks show genes and enhancers in ovary as described in Hnisz et al.38. Positions of SNPs for which imputation r2 < 0.3 and/or minor allele frequency < 0.005 are shown in the bottom track as ‘untyped’ SNPs.

Supplementary Figure 7 Ovarian cancer susceptibility loci at chromosome 6 and chromosome 9.

The Manhattan plot depicts the strength of association between all imputed and genotyped SNPs across the regions at chromosome 6 (a) and chromosome 9 (b). The dotted line represents the genome-wide significance level 5 × 10−8. FAIRE-seq data revealing potential regulatory regions in ovarian and fallopian tube cells are depicted as black bars. Additional tracks show genes and enhancers in ovary as described in Hnisz et al.38. Positions of SNPs for which imputation r2 < 0.3 and/or minor allele frequency < 0.005 are shown in the bottom track as ‘untyped’ SNPs.

Supplementary Figure 8 Ovarian cancer susceptibility locus at chromosome 17.

The Manhattan plot depicts the strength of association between all imputed and genotyped SNPs across the regions at chromosome 17. The dotted line represents the genome-wide significance level 5 × 10−8. FAIRE-seq data revealing potential regulatory regions in ovarian and fallopian tube cells are depicted as black bars. Additional tracks show genes and enhancers in ovary as described in Hnisz et al.38. Positions of SNPs for which imputation r2 < 0.3 and/or minor allele frequency < 0.005 are shown in the bottom track as ‘untyped’ SNPs.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8, Supplementary Tables 1–12 and Supplementary Note. (PDF 2825 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kuchenbaecker, K., Ramus, S., Tyrer, J. et al. Identification of six new susceptibility loci for invasive epithelial ovarian cancer. Nat Genet 47, 164–171 (2015). https://doi.org/10.1038/ng.3185

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3185

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer