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:

Five endometrial cancer risk loci identified through genome-wide association analysis

Abstract

We conducted a meta-analysis of three endometrial cancer genome-wide association studies (GWAS) and two follow-up phases totaling 7,737 endometrial cancer cases and 37,144 controls of European ancestry. Genome-wide imputation and meta-analysis identified five new risk loci of genome-wide significance at likely regulatory regions on chromosomes 13q22.1 (rs11841589, near KLF5), 6q22.31 (rs13328298, in LOC643623 and near HEY2 and NCOA7), 8q24.21 (rs4733613, telomeric to MYC), 15q15.1 (rs937213, in EIF2AK4, near BMF) and 14q32.33 (rs2498796, in AKT1, near SIVA1). We also found a second independent 8q24.21 signal (rs17232730). Functional studies of the 13q22.1 locus showed that rs9600103 (pairwise r2 = 0.98 with rs11841589) is located in a region of active chromatin that interacts with the KLF5 promoter region. The rs9600103[T] allele that is protective in endometrial cancer suppressed gene expression in vitro, suggesting that regulation of the expression of KLF5, a gene linked to uterine development, is implicated in tumorigenesis. These findings provide enhanced insight into the genetic and biological basis of endometrial cancer.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: Manhattan plot from endometrial cancer meta-analysis.
Figure 2: Forest plots for new endometrial cancer risk loci.
Figure 3: Regional association plots for the five new loci associated with endometrial cancer.
Figure 4: The 13q22.1 endometrial cancer susceptibility locus.

Similar content being viewed by others

References

  1. Siegel, R., Ma, J., Zou, Z. & Jemal, A. Cancer statistics, 2014. CA Cancer J. Clin. 64, 9–29 (2014).

    PubMed  Google Scholar 

  2. Ferlay, J. et al. Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012. Eur. J. Cancer 49, 1374–1403 (2013).

    CAS  PubMed  Google Scholar 

  3. Gruber, S.B., Thompson, W.D. & Cancer and Steroid Hormone Study Group. A population-based study of endometrial cancer and familial risk in younger women. Cancer Epidemiol. Biomarkers Prev. 5, 411–417 (1996).

    CAS  PubMed  Google Scholar 

  4. Win, A.K., Reece, J.C. & Ryan, S. Family history and risk of endometrial cancer: a systematic review and meta-analysis. Obstet. Gynecol. 125, 89–98 (2015).

    Article  PubMed  Google Scholar 

  5. Barrow, E., Hill, J. & Evans, D.G. Cancer risk in Lynch syndrome. Fam. Cancer 12, 229–240 (2013).

    Article  CAS  PubMed  Google Scholar 

  6. Church, D.N. et al. DNA polymerase ɛ and δ exonuclease domain mutations in endometrial cancer. Hum. Mol. Genet. 22, 2820–2828 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Palles, C. et al. Germline mutations affecting the proofreading domains of POLE and POLD1 predispose to colorectal adenomas and carcinomas. Nat. Genet. 45, 136–144 (2013).

    Article  CAS  PubMed  Google Scholar 

  8. Painter, J.N. et al. Fine-mapping of the HNF1B multicancer locus identifies candidate variants that mediate endometrial cancer risk. Hum. Mol. Genet. 24, 1478–1492 (2015).

    Article  CAS  PubMed  Google Scholar 

  9. Thompson, D.J. et al. CYP19A1 fine-mapping and Mendelian randomization: estradiol is causal for endometrial cancer. Endocr. Relat. Cancer 23, 77–91 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Spurdle, A.B. et al. Genome-wide association study identifies a common variant associated with risk of endometrial cancer. Nat. Genet. 43, 451–454 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Tenesa, A. et al. Genome-wide association scan identifies a colorectal cancer susceptibility locus on 11q23 and replicates risk loci at 8q24 and 18q21. Nat. Genet. 40, 631–637 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Tomlinson, I. et al. A genome-wide association scan of tag SNPs identifies a susceptibility variant for colorectal cancer at 8q24.21. Nat. Genet. 39, 984–988 (2007).

    Article  CAS  PubMed  Google Scholar 

  13. Wellcome Trust Case Control Consortium. Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature 447, 661–678 (2007).

  14. Eeles, R.A. et al. Identification of 23 new prostate cancer susceptibility loci using the iCOGS custom genotyping array. Nat. Genet. 45, 385–391, e1–e2 (2013).

    Article  CAS  PubMed  Google Scholar 

  15. Michailidou, K. et al. Large-scale genotyping identifies 41 new loci associated with breast cancer risk. Nat. Genet. 45, 353–361, e1–e2 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Pharoah, P.D. et al. GWAS meta-analysis and replication identifies three new susceptibility loci for ovarian cancer. Nat. Genet. 45, 362–370, e1–e2 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Sakoda, L.C., Jorgenson, E. & Witte, J.S. Turning of COGS moves forward findings for hormonally mediated cancers. Nat. Genet. 45, 345–348 (2013).

    Article  CAS  PubMed  Google Scholar 

  18. De Vivo, I. et al. Genome-wide association study of endometrial cancer in E2C2. Hum. Genet. 133, 211–224 (2014).

    Article  CAS  PubMed  Google Scholar 

  19. Setiawan, V.W. et al. Two estrogen-related variants in CYP19A1 and endometrial cancer risk: a pooled analysis in the Epidemiology of Endometrial Cancer Consortium. Cancer Epidemiol. Biomarkers Prev. 18, 242–247 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. GTEx Consortium. The Genotype-Tissue Expression (GTEx) project. Nat. Genet. 45, 580–585 (2013).

  21. Cancer Genome Atlas Research Network. Integrated genomic characterization of endometrial carcinoma. Nature 497, 67–73 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Katoh, M. & Katoh, M. Integrative genomic analyses on HES/HEY family: Notch-independent HES1, HES3 transcription in undifferentiated ES cells, and Notch-dependent HES1, HES5, HEY1, HEY2, HEYL transcription in fetal tissues, adult tissues, or cancer. Int. J. Oncol. 31, 461–466 (2007).

    CAS  PubMed  Google Scholar 

  23. Shao, W., Halachmi, S. & Brown, M. ERAP140, a conserved tissue-specific nuclear receptor coactivator. Mol. Cell. Biol. 22, 3358–3372 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Kiemeney, L.A. et al. Sequence variant on 8q24 confers susceptibility to urinary bladder cancer. Nat. Genet. 40, 1307–1312 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Rothman, N. et al. A multi-stage genome-wide association study of bladder cancer identifies multiple susceptibility loci. Nat. Genet. 42, 978–984 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Easton, D.F. et al. Genome-wide association study identifies novel breast cancer susceptibility loci. Nature 447, 1087–1093 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Whiffin, N. et al. Identification of susceptibility loci for colorectal cancer in a genome-wide meta-analysis. Hum. Mol. Genet. 23, 4729–4737 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. 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 

  29. Eeles, R.A. et al. Identification of seven new prostate cancer susceptibility loci through a genome-wide association study. Nat. Genet. 41, 1116–1121 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Gudmundsson, J. et al. Genome-wide association and replication studies identify four variants associated with prostate cancer susceptibility. Nat. Genet. 41, 1122–1126 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Huppi, K., Pitt, J.J., Wahlberg, B.M. & Caplen, N.J. The 8q24 gene desert: an oasis of non-coding transcriptional activity. Front. Genet. 3, 69 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Carvajal-Carmona, L.G. et al. Candidate locus analysis of the TERTCLPTM1L cancer risk region on chromosome 5p15 identifies multiple independent variants associated with endometrial cancer risk. Hum. Genet. 134, 231–245 (2015).

    Article  CAS  PubMed  Google Scholar 

  33. Berlanga, J.J., Santoyo, J. & De Haro, C. Characterization of a mammalian homolog of the GCN2 eukaryotic initiation factor 2α kinase. Eur. J. Biochem. 265, 754–762 (1999).

    Article  CAS  PubMed  Google Scholar 

  34. Uhlén, M. et al. Tissue-based map of the human proteome. Science 347, 1260419 (2015).

    Article  CAS  PubMed  Google Scholar 

  35. Cantley, L.C. The phosphoinositide 3-kinase pathway. Science 296, 1655–1657 (2002).

    Article  CAS  PubMed  Google Scholar 

  36. Slomovitz, B.M. & Coleman, R.L. The PI3K/AKT/mTOR pathway as a therapeutic target in endometrial cancer. Clin. Cancer Res. 18, 5856–5864 (2012).

    Article  CAS  PubMed  Google Scholar 

  37. Cohen, Y. et al. AKT1 pleckstrin homology domain E17K activating mutation in endometrial carcinoma. Gynecol. Oncol. 116, 88–91 (2010).

    Article  CAS  PubMed  Google Scholar 

  38. Salvesen, H.B. et al. Integrated genomic profiling of endometrial carcinoma associates aggressive tumors with indicators of PI3 kinase activation. Proc. Natl. Acad. Sci. USA 106, 4834–4839 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Shoji, K. et al. The oncogenic mutation in the pleckstrin homology domain of AKT1 in endometrial carcinomas. Br. J. Cancer 101, 145–148 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Du, W. et al. Suppression of p53 activity by Siva1. Cell Death Differ. 16, 1493–1504 (2009).

    Article  CAS  PubMed  Google Scholar 

  41. Wang, X. et al. Siva1 inhibits p53 function by acting as an ARF E3 ubiquitin ligase. Nat. Commun. 4, 1551 (2013).

    Article  PubMed  CAS  Google Scholar 

  42. Li, N. et al. Siva1 suppresses epithelial–mesenchymal transition and metastasis of tumor cells by inhibiting stathmin and stabilizing microtubules. Proc. Natl. Acad. Sci. USA 108, 12851–12856 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Dun, B. et al. Mycophenolic acid inhibits migration and invasion of gastric cancer cells via multiple molecular pathways. PLoS One 8, e81702 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Davis, H. et al. FBXW7 mutations typically found in human cancers are distinct from null alleles and disrupt lung development. J. Pathol. 224, 180–189 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Mutter, G.L. et al. Global expression changes of constitutive and hormonally regulated genes during endometrial neoplastic transformation. Gynecol. Oncol. 83, 177–185 (2001).

    Article  CAS  PubMed  Google Scholar 

  46. Shi, H., Zhang, Z., Wang, X., Liu, S. & Teng, C.T. Isolation and characterization of a gene encoding human Kruppel-like factor 5 (IKLF): binding to the CAAT/GT box of the mouse lactoferrin gene promoter. Nucleic Acids Res. 27, 4807–4815 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Simmen, R.C. et al. The emerging role of Krüppel-like factors in endocrine-responsive cancers of female reproductive tissues. J. Endocrinol. 204, 223–231 (2010).

    Article  CAS  PubMed  Google Scholar 

  48. Nandan, M.O. et al. Krüppel-like factor 5 mediates cellular transformation during oncogenic KRAS-induced intestinal tumorigenesis. Gastroenterology 134, 120–130 (2008).

    Article  CAS  PubMed  Google Scholar 

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

  50. ENCODE Project Consortium. Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot project. Nature 447, 799–816 (2007).

  51. Rao, S.S. et al. A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell 159, 1665–1680 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Wang, Y., Li, X. & Hu, H. H3K4me2 reliably defines transcription factor binding regions in different cells. Genomics 103, 222–228 (2014).

    Article  CAS  PubMed  Google Scholar 

  53. Cheng, T.H. et al. Meta-analysis of genome-wide association studies identifies common susceptibility polymorphisms for colorectal and endometrial cancer near SH2B3 and TSHZ1 . Sci. Rep. 5, 17369 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. O'Mara, T.A. et al. Comprehensive genetic assessment of the ESR1 locus identifies a risk region for endometrial cancer. Endocr. Relat. Cancer 22, 851–861 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Power, C. & Elliott, J. Cohort profile: 1958 British birth cohort (National Child Development Study). Int. J. Epidemiol. 35, 34–41 (2006).

    Article  PubMed  Google Scholar 

  56. McEvoy, M. et al. Cohort profile: the Hunter Community Study. Int. J. Epidemiol. 39, 1452–1463 (2010).

    Article  PubMed  Google Scholar 

  57. McGregor, B. et al. Genetic and environmental contributions to size, color, shape, and other characteristics of melanocytic naevi in a sample of adolescent twins. Genet. Epidemiol. 16, 40–53 (1999).

    Article  CAS  PubMed  Google Scholar 

  58. Teo, Y.Y. et al. A genotype calling algorithm for the Illumina BeadArray platform. Bioinformatics 23, 2741–2746 (2007).

    Article  CAS  PubMed  Google Scholar 

  59. Price, A.L. et al. Principal components analysis corrects for stratification in genome-wide association studies. Nat. Genet. 38, 904–909 (2006).

    Article  CAS  PubMed  Google Scholar 

  60. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Aulchenko, Y.S., Ripke, S., Isaacs, A. & van Duijn, C.M. GenABEL: an R library for genome-wide association analysis. Bioinformatics 23, 1294–1296 (2007).

    Article  CAS  PubMed  Google Scholar 

  62. Clayton, D. & Leung, H.T. An R package for analysis of whole-genome association studies. Hum. Hered. 64, 45–51 (2007).

    Article  PubMed  Google Scholar 

  63. Howie, B., Marchini, J. & Stephens, M. Genotype imputation with thousands of genomes. G3 (Bethesda) 1, 457–470 (2011).

    Article  Google Scholar 

  64. Delaneau, O., Zagury, J.F. & Marchini, J. Improved whole-chromosome phasing for disease and population genetic studies. Nat. Methods 10, 5–6 (2013).

    Article  CAS  PubMed  Google Scholar 

  65. Timpson, N.J. et al. A rare variant in APOC3 is associated with plasma triglyceride and VLDL levels in Europeans. Nat. Commun. 5, 4871 (2014).

    Article  CAS  PubMed  Google Scholar 

  66. 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).

    Article  CAS  PubMed  Google Scholar 

  67. Pruim, R.J. et al. LocusZoom: regional visualization of genome-wide association scan results. Bioinformatics 26, 2336–2337 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Mägi, R. & Morris, A.P. GWAMA: software for genome-wide association meta-analysis. BMC Bioinformatics 11, 288 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  69. Higgins, J.P. & Thompson, S.G. Quantifying heterogeneity in a meta-analysis. Stat. Med. 21, 1539–1558 (2002).

    Article  PubMed  Google Scholar 

  70. Huedo-Medina, T.B., Sánchez-Meca, J., Marín-Martínez, F. & Botella, J. Assessing heterogeneity in meta-analysis: Q statistic or I2 index? Psychol. Methods 11, 193–206 (2006).

    Article  PubMed  Google Scholar 

  71. Ward, L.D. & Kellis, M. HaploReg: a resource for exploring chromatin states, conservation, and regulatory motif alterations within sets of genetically linked variants. Nucleic Acids Res. 40, D930–D934 (2012).

    Article  CAS  PubMed  Google Scholar 

  72. Boyle, A.P. et al. Annotation of functional variation in personal genomes using RegulomeDB. Genome Res. 22, 1790–1797 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  74. Corradin, O. et al. Combinatorial effects of multiple enhancer variants in linkage disequilibrium dictate levels of gene expression to confer susceptibility to common traits. Genome Res. 24, 1–13 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Livak, K.J. & Schmittgen, T.D. Analysis of relative gene expression data using real-time quantitative PCR and the 2(−ΔΔC T ) method. Methods 25, 402–408 (2001).

    Article  CAS  PubMed  Google Scholar 

  76. Giresi, P.G., Kim, J., McDaniell, R.M., Iyer, V.R. & Lieb, J.D. FAIRE (formaldehyde-assisted isolation of regulatory elements) isolates active regulatory elements from human chromatin. Genome Res. 17, 877–885 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Ghoussaini, M. et al. Evidence that breast cancer risk at the 2q35 locus is mediated through IGFBP5 regulation. Nat. Commun. 4, 4999 (2014).

    Article  PubMed  CAS  Google Scholar 

  78. Lewis, A. et al. A polymorphic enhancer near GREM1 influences bowel cancer risk through differential CDX2 and TCF7L2 binding. Cell Rep. 8, 983–990 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the many individuals who participated in this study and the numerous institutions and their staff that supported recruitment, detailed in full in the Supplementary Note.

The iCOGS endometrial cancer analysis was supported by an NHMRC project grant (1031333) to A.B.S., D.F.E. and A.M.D. A.B.S., P.M.W., G.W.M. and D.R.N. are supported by the NHMRC Fellowship scheme. A.M.D. was supported by the Joseph Mitchell Trust. I.T. is supported by Cancer Research UK and the Oxford Comprehensive Biomedical Research Centre. T.H.T.C. is supported by the Rhodes Trust and the Nuffield Department of Medicine. Funding for iCOGS infrastructure came from the European Community's Seventh Framework Programme under grant agreement 223175 (HEALTH-F2-2009-223175) (COGS), Cancer Research UK (C1287/A10118, C1287/A10710, C12292/A11174, C1281/A12014, C5047/A8384, C5047/A15007, C5047/A10692 and C8197/A16565), the US National Institutes of Health (R01 CA128978, U19 CA148537, U19 CA148065 and U19 CA148112), the US Department of Defense (W81XWH-10-1-0341), the Canadian Institutes of Health Research (CIHR) for the CIHR Team in Familial Risks of Breast Cancer, the Susan G. Komen Foundation for the Cure, the Breast Cancer Research Foundation and the Ovarian Cancer Research Fund.

ANECS recruitment was supported by project grants from the NHMRC (339435), Cancer Council Queensland (4196615) and Cancer Council Tasmania (403031 and 457636). SEARCH recruitment was funded by a programme grant from Cancer Research UK (C490/A10124). Stage 1 and stage 2 case genotyping was supported by the NHMRC (552402 and 1031333). Control data were generated by the WTCCC, and a full list of the investigators who contributed to the generation of the data is available from the WTCCC website. We acknowledge use of DNA from the British 1958 Birth Cohort collection, funded by UK Medical Research Council grant G0000934 and Wellcome Trust grant 068545/Z/02; funding for this project was provided by the Wellcome Trust under award 085475. NSECG was supported by the European Union's Framework Programme 7 CHIBCHA grant and Wellcome Trust Centre for Human Genetics Core Grant 090532/Z/09Z, and CORGI was funded by Cancer Research UK. Recruitment of the QIMR Berghofer controls was supported by the NHMRC. The University of Newcastle, the Gladys M. Brawn Senior Research Fellowship scheme, the Vincent Fairfax Family Foundation, the Hunter Medical Research Institute and the Hunter Area Pathology Service all contributed toward the costs of establishing HCS.

The Bavarian Endometrial Cancer Study (BECS) was partly funded by the ELAN fund of the University of Erlangen. The Hannover–Jena Endometrial Cancer Study was partly supported by the Rudolf Bartling Foundation. The Leuven Endometrium Study (LES) was supported by the Verelst Foundation for Endometrial Cancer. The Mayo Endometrial Cancer Study (MECS) and Mayo controls (MAY) were supported by grants from the National Cancer Institute of the US Public Health Service (R01 CA122443, P30 CA15083 and P50 CA136393), the Fred C. and Katherine B. Andersen Foundation, the Mayo Foundation and the Ovarian Cancer Research Fund with support of the Smith family, in memory of Kathryn Sladek Smith. MoMaTEC received financial support from a Helse Vest Grant, the University of Bergen, the Melzer Foundation, the Norwegian Cancer Society (Harald Andersens legat), the Research Council of Norway and Haukeland University Hospital. The Newcastle Endometrial Cancer Study (NECS) acknowledges contributions from the University of Newcastle, the NBN Children's Cancer Research Group, Jennie Thomas and the Hunter Medical Research Institute. RENDOCAS was supported through the regional agreement on medical training and clinical research (ALF) between the Stockholm County Council and Karolinska Institutet (20110222, 20110483, 20110141 and DF 07015), Swedish Labor Market Insurance (100069) and the Swedish Cancer Society (11 0439). The Cancer Hormone Replacement Epidemiology in Sweden study (CAHRES; formerly called the Singapore and Swedish Breast/Endometrial Cancer study, SASBAC) was supported by funding from the Agency for Science, Technology and Research of Singapore (A*STAR), the US National Institutes of Health and the Susan G. Komen Breast Cancer Foundation.

BCAC is funded by Cancer Research UK (C1287/A10118 and C1287/A12014). 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) and the UK National Institute for Health Research Biomedical Research Centres at the University of Cambridge.

Additional funding for individual control groups is detailed in the Supplementary Note.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

A.B.S., D.F.E., A.M.D., G.W.M. and P.M.W. obtained funding for the study. A.B.S. and D.F.E. designed the study. T.H.T.C., D.J.T., T.A.O'M., J.N.P., D.M.G., I.T. and A.B.S. drafted the manuscript. T.H.T.C. and D.J.T. conducted statistical analyses and genotype imputation. T.A.O'M., D.M.G., M.J.L., S.H.Y. and J.W. conducted bioinformatic analyses. T.A.O'M. conducted eQTL analyses. S.F., A. Lewis, J.D.F., L.F.-M., D.C. and S.L.E. performed functional assays. T.H.T.C., T.A.O'M. and J.N.P. performed additional genotyping by KASPar and Fluidigm. T.A.O'M. coordinated the overall stage 2 genotyping and associated data management. J. Dennis, J.P.T. and K.M. coordinated quality control and data cleaning for the iCOGS control data sets. A.B.S. and T.A.O'M. coordinated the ANECS stage 1 genotyping. A.M.D., S.A. and C.S.H. coordinated the SEARCH stage 1 genotyping. I.T. and CHIBCHA funded and implemented the NSECG GWAS. I.T., L.M., M.G. and S.H. coordinated NSECG and collation of CORGI control GWAS data. A.B.S. and P.M.W. coordinated ANECS. R.J.S., M. McEvoy, J.A. and E.G.H. coordinated collation of GWAS data for HCS. N.G.M., G.W.M., D.R.N. and A.K.H. coordinated collation of GWAS data for the QIMR controls. P.D.P.P., D.F.E. and M.S. coordinated SEARCH. M.K.B. and Q.W. provided data management support for BCAC. The following authors designed and coordinated the baseline studies and/or extraction of questionnaire and clinical information for studies: P.A.F., M.W.B., A.H., A.B.E., T.D., P. Hillemanns, M. Dürst, I.R., D.L., S.S., H.Z., F.A., J. Depreeuw, S.C.D., E.L.G., B.L.F., S.J.W., H.B.S., J.T., T.S.N., H.M.J.W., R.J.S., K.A., T.P., G.O., T.L., M. Mints, E.T., P. Hall, K.C., J.L., H.D., M. Dunlop, R.H., C.P., J.L.H., J.P., A.J.S., B.B., H. Brenner, A.M., H. Brauch, A. Lindblom, J.C.-C., F.J.C., G.G.G., V.N.K., A.C. and J.M.C. All authors provided critical review of the manuscript.

Corresponding authors

Correspondence to Douglas F Easton, Ian Tomlinson or Amanda B Spurdle.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

A list of members appears in the Supplementary Note.

A list of members appears in the Supplementary Note.

A list of members appears in the Supplementary Note.

A list of members appears in the Supplementary Note.

A list of members appears in the Supplementary Note.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 and Supplementary Note. (PDF 3434 kb)

Supplementary Table 1

Endometrial cancer case and control sample sets. (XLSX 14 kb)

Supplementary Table 2

Meta-analysis after regional imputation for risk loci with P < 1 × 10−5 identified by meta-analysis of GWAS and iCOGS data sets. (XLSX 12 kb)

Supplementary Table 3

Overall meta-analysis including additional genotyping from phase 2. (XLSX 10 kb)

Supplementary Table 4

Genotyping concordance rates for different platforms in quality control duplicates. (XLSX 8 kb)

Supplementary Table 5

Endometrial tissue eQTLs: association between GWAS risk locus genotypes and transcript levels of nearby genes. (XLSX 11 kb)

Supplementary Table 6

Functional annotation of SNPs in LD with GWAS risk loci (r2 >0.7 in 1000 Genomes Project EUR) from HaploReg, RegulomeDB and ENCODE. (XLSX 35 kb)

Supplementary Table 7

Pairwise t-test P values for 13q22 luciferase assays. (XLSX 8 kb)

Supplementary Table 8

Primers and oligonucleotides used in experimental procedures. (XLSX 11 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cheng, T., Thompson, D., O'Mara, T. et al. Five endometrial cancer risk loci identified through genome-wide association analysis. Nat Genet 48, 667–674 (2016). https://doi.org/10.1038/ng.3562

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3562

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