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:

Exome sequencing identifies recurrent SPOP, FOXA1 and MED12 mutations in prostate cancer

Abstract

Prostate cancer is the second most common cancer in men worldwide and causes over 250,000 deaths each year1. Overtreatment of indolent disease also results in significant morbidity2. Common genetic alterations in prostate cancer include losses of NKX3.1 (8p21)3,4 and PTEN (10q23)5,6, gains of AR (the androgen receptor gene)7,8 and fusion of ETS family transcription factor genes with androgen-responsive promoters9,10,11. Recurrent somatic base-pair substitutions are believed to be less contributory in prostate tumorigenesis12,13 but have not been systematically analyzed in large cohorts. Here, we sequenced the exomes of 112 prostate tumor and normal tissue pairs. New recurrent mutations were identified in multiple genes, including MED12 and FOXA1. SPOP was the most frequently mutated gene, with mutations involving the SPOP substrate-binding cleft in 6–15% of tumors across multiple independent cohorts. Prostate cancers with mutant SPOP lacked ETS family gene rearrangements and showed a distinct pattern of genomic alterations. Thus, SPOP mutations may define a new molecular subtype of prostate 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: Significantly mutated genes in aggressive primary prostate cancer.
Figure 2: Recurrent somatic alterations in FOXA1 and MED12.
Figure 3: Structural and functional studies of recurrent SPOP alterations in prostate cancer.
Figure 4: SPOP mutation defines a distinct genetic subclass of prostate cancer.

Similar content being viewed by others

References

  1. Jemal, A. et al. Global cancer statistics. CA Cancer J. Clin. 61, 69–90 (2011).

    Article  Google Scholar 

  2. Daskivich, T.J. et al. Overtreatment of men with low-risk prostate cancer and significant comorbidity. Cancer 117, 2058–2066 (2011).

    Article  Google Scholar 

  3. He, W.W. et al. A novel human prostate-specific, androgen-regulated homeobox gene (NKX3.1) that maps to 8p21, a region frequently deleted in prostate cancer. Genomics 43, 69–77 (1997).

    Article  CAS  Google Scholar 

  4. Bhatia-Gaur, R. et al. Roles for Nkx3.1 in prostate development and cancer. Genes Dev. 13, 966–977 (1999).

    Article  CAS  Google Scholar 

  5. Li, J. et al. PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer. Science 275, 1943–1947 (1997).

    Article  CAS  Google Scholar 

  6. Cairns, P. et al. Frequent inactivation of PTEN/MMAC1 in primary prostate cancer. Cancer Res. 57, 4997–5000 (1997).

    CAS  PubMed  Google Scholar 

  7. Linja, M.J. & Visakorpi, T. Alterations of androgen receptor in prostate cancer. J. Steroid Biochem. Mol. Biol. 92, 255–264 (2004).

    Article  CAS  Google Scholar 

  8. Visakorpi, T. et al. In vivo amplification of the androgen receptor gene and progression of human prostate cancer. Nat. Genet. 9, 401–406 (1995).

    Article  CAS  Google Scholar 

  9. Perner, S. et al. TMPRSS2:ERG fusion–associated deletions provide insight into the heterogeneity of prostate cancer. Cancer Res. 66, 8337–8341 (2006).

    Article  CAS  Google Scholar 

  10. Tomlins, S.A. et al. Distinct classes of chromosomal rearrangements create oncogenic ETS gene fusions in prostate cancer. Nature 448, 595–599 (2007).

    Article  CAS  Google Scholar 

  11. Tomlins, S.A. et al. Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science 310, 644–648 (2005).

    Article  CAS  Google Scholar 

  12. Taylor, B.S. et al. Integrative genomic profiling of human prostate cancer. Cancer Cell 18, 11–22 (2010).

    Article  CAS  Google Scholar 

  13. Kumar, A. et al. Exome sequencing identifies a spectrum of mutation frequencies in advanced and lethal prostate cancers. Proc. Natl. Acad. Sci. U.S.A. 108, 17087–17092 (2011).

    Article  CAS  Google Scholar 

  14. Berger, M.F. et al. The genomic complexity of primary human prostate cancer. Nature 470, 214–220 (2011).

    Article  CAS  Google Scholar 

  15. Nagai, Y. et al. Identification of a novel nuclear speckle-type protein, SPOP. FEBS Lett. 418, 23–26 (1997).

    Article  CAS  Google Scholar 

  16. Zhuang, M. et al. Structures of SPOP-substrate complexes: insights into molecular architectures of BTB-Cul3 ubiquitin ligases. Mol. Cell 36, 39–50 (2009).

    Article  CAS  Google Scholar 

  17. Kan, Z. et al. Diverse somatic mutation patterns and pathway alterations in human cancers. Nature 466, 869–873 (2010).

    Article  CAS  Google Scholar 

  18. Majumder, P.K. et al. A prostatic intraepithelial neoplasia–dependent p27Kip1 checkpoint induces senescence and inhibits cell proliferation and cancer progression. Cancer Cell 14, 146–155 (2008).

    Article  CAS  Google Scholar 

  19. Kibel, A.S. et al. CDKN1A and CDKN1B polymorphisms and risk of advanced prostate carcinoma. Cancer Res. 63, 2033–2036 (2003).

    CAS  PubMed  Google Scholar 

  20. Gao, N. et al. Forkhead box A1 regulates prostate ductal morphogenesis and promotes epithelial cell maturation. Development 132, 3431–3443 (2005).

    Article  CAS  Google Scholar 

  21. Zhang, C. et al. Definition of a FoxA1 cistrome that is crucial for G1 to S-phase cell-cycle transit in castration-resistant prostate cancer. Cancer Res. 71, 6738–6748 (2011).

    Article  CAS  Google Scholar 

  22. Gao, N. et al. The role of hepatocyte nuclear factor–3α (Forkhead Box A1) and androgen receptor in transcriptional regulation of prostatic genes. Mol. Endocrinol. 17, 1484–1507 (2003).

    Article  CAS  Google Scholar 

  23. Williamson, E.A. et al. BRCA1 and FOXA1 proteins coregulate the expression of the cell cycle–dependent kinase inhibitor p27Kip1. Oncogene 25, 1391–1399 (2006).

    Article  CAS  Google Scholar 

  24. Clark, K.L., Halay, E.D., Lai, E. & Burley, S.K. Co-crystal structure of the HNF-3/fork head DNA-recognition motif resembles histone H5. Nature 364, 412–420 (1993).

    Article  CAS  Google Scholar 

  25. Zhou, R. et al. SOX9 interacts with a component of the human thyroid hormone receptor–associated protein complex. Nucleic Acids Res. 30, 3245–3252 (2002).

    Article  CAS  Google Scholar 

  26. Wang, Q., Sharma, D., Ren, Y. & Fondell, J.D. A coregulatory role for the TRAP-mediator complex in androgen receptor–mediated gene expression. J. Biol. Chem. 277, 42852–42858 (2002).

    Article  CAS  Google Scholar 

  27. Donner, A.J., Szostek, S., Hoover, J.M. & Espinosa, J.M. CDK8 is a stimulus-specific positive coregulator of p53 target genes. Mol. Cell 27, 121–133 (2007).

    Article  CAS  Google Scholar 

  28. Mäkinen, N. et al. MED12, the mediator complex subunit 12 gene, is mutated at high frequency in uterine leiomyomas. Science 334, 252–255 (2011).

    Article  Google Scholar 

  29. Liu, J. et al. Analysis of Drosophila segmentation network identifies a JNK pathway factor overexpressed in kidney cancer. Science 323, 1218–1222 (2009).

    Article  CAS  Google Scholar 

  30. Wang, C., Pan, Y. & Wang, B. Suppressor of fused and Spop regulate the stability, processing and function of Gli2 and Gli3 full-length activators but not their repressors. Development 137, 2001–2009 (2010).

    Article  CAS  Google Scholar 

  31. Li, C. et al. Tumor-suppressor role for the SPOP ubiquitin ligase in signal-dependent proteolysis of the oncogenic co-activator SRC-3/AIB1. Oncogene 30, 4350–4364 (2011).

    Article  CAS  Google Scholar 

  32. Mosquera, J.M. et al. Prevalence of TMPRSS2-ERG fusion prostate cancer among men undergoing prostate biopsy in the United States. Clin. Cancer Res. 15, 4706–4711 (2009).

    Article  CAS  Google Scholar 

  33. Demichelis, F. et al. Distinct genomic aberrations associated with ERG rearranged prostate cancer. Genes Chromosom. Cancer 48, 366–380 (2009).

    Article  CAS  Google Scholar 

  34. Lapointe, J. et al. Genomic profiling reveals alternative genetic pathways of prostate tumorigenesis. Cancer Res. 67, 8504–8510 (2007).

    Article  CAS  Google Scholar 

  35. Yang, J.Y. & Hung, M.C. A new fork for clinical application: targeting forkhead transcription factors in cancer. Clin. Cancer Res. 15, 752–757 (2009).

    Article  CAS  Google Scholar 

  36. Mandelbaum, J. et al. BLIMP1 is a tumor suppressor gene frequently disrupted in activated B cell–like diffuse large B cell lymphoma. Cancer Cell 18, 568–579 (2010).

    Article  CAS  Google Scholar 

  37. Fisher, S. et al. A scalable, fully automated process for construction of sequence-ready human exome targeted capture libraries. Genome Biol. 12, R1 (2011).

    Article  Google Scholar 

  38. Stransky, N. et al. The mutational landscape of head and neck squamous cell carcinoma. Science 333, 1157–1160 (2011).

    Article  CAS  Google Scholar 

  39. Chapman, M.A. et al. Initial genome sequencing and analysis of multiple myeloma. Nature 471, 467–472 (2011).

    Article  CAS  Google Scholar 

  40. Reich, M. et al. GenePattern 2.0. Nat. Genet. 38, 500–501 (2006).

    Article  CAS  Google Scholar 

  41. Cibulskis, K. et al. ContEst: estimating cross-contamination of human samples in next-generation sequencing data. Bioinformatics 27, 2601–2602 (2011).

    Article  CAS  Google Scholar 

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

  43. Robinson, J.T. et al. Integrative genomics viewer. Nat. Biotechnol. 29, 24–26 (2011).

    Article  CAS  Google Scholar 

  44. Fujita, P.A. et al. The UCSC Genome Browser database: update 2011. Nucleic Acids Res. 39, D876–D882 (2011).

    Article  CAS  Google Scholar 

  45. Kozomara, A. & Griffiths-Jones, S. miRBase: integrating microRNA annotation and deep-sequencing data. Nucleic Acids Res. 39, D152–D157 (2011).

    Article  CAS  Google Scholar 

  46. Sherry, S.T. et al. dbSNP: the NCBI database of genetic variation. Nucleic Acids Res. 29, 308–311 (2001).

    Article  CAS  Google Scholar 

  47. Griffith, O.L. et al. ORegAnno: an open-access community-driven resource for regulatory annotation. Nucleic Acids Res. 36, D107–D113 (2008).

    Article  CAS  Google Scholar 

  48. UniProt Consortium. Ongoing and future developments at the Universal Protein Resource. Nucleic Acids Res. 39, D214–D219 (2011).

  49. Forbes, S.A. et al. COSMIC: mining complete cancer genomes in the Catalogue of Somatic Mutations in Cancer. Nucleic Acids Res. 39, D945–D950 (2011).

    Article  CAS  Google Scholar 

  50. Carter, S.L. et al. Absolute quantification of somatic DNA alterations in human cancer. Nat. Biotechnol. published online, doi:10.1038/nbt.2203 (29 April 2012).

  51. Pflueger, D. et al. Discovery of non-ETS gene fusions in human prostate cancer using next-generation RNA sequencing. Genome Res. 21, 56–67 (2011).

    Article  CAS  Google Scholar 

  52. Espina, V. et al. Laser-capture microdissection. Nat. Protoc. 1, 586–603 (2006).

    Article  CAS  Google Scholar 

  53. Svensson, M.A. et al. Testing mutual exclusivity of ETS rearranged prostate cancer. Lab. Invest. 91, 404–412 (2011).

    Article  CAS  Google Scholar 

  54. Park, K. et al. Antibody-based detection of ERG rearrangement–positive prostate cancer. Neoplasia 12, 590–598 (2010).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We are grateful for the assistance of members of the Broad Institute Biological Samples Platform, Genetic Analysis Platform and Genome Sequencing Platform. We thank S. Banerjee for computational assistance, R. Kim and R. Leung for their critical contributions to the Weill Cornell Prostate Cancer Tumor Bank, P. Schraml, S. Dettwiler and M. Storz for assistance with the University Hospital Zurich cohort and biobank and the members of the University of Washington Rapid Autopsy Program. We thank the University of Michigan Prostate Cancer Special Program of Research Excellence (SPORE) (K. Pienta) for sample contribution to this study. We are also grateful to the individuals with prostate cancer and the families who contributed to these studies. This work was supported by the US National Human Genome Research Institute (NHGRI) Large Scale Sequencing Program (U54 HG003067 to the Broad Institute, E.S.L.), the Kohlberg Foundation (L.A.G.), the Starr Cancer Consortium (M.A.R., F.D. and L.A.G), the Prostate Cancer Foundation (M.A.R.), US Department of Defense Synergy Awards (PC101020 to F.D., L.A.G. and M.A.R. and PC093372 to T.W. and P.S.N.), the Dana-Farber/Harvard Cancer Center Prostate Cancer SPORE (US National Institutes of Health (NIH) P50 CA090381), a New Investigator Award (PC094516 to F.D.), the US National Cancer Institute, Early Detection Research Network (U01CA111275 and NCI EDRN to F.D. and M.A.R.), the US National Cancer Institute (R01 CA125612 to F.D. and M.A.R.), the Pacific Northwest Prostate Cancer SPORE (P50CA097186 to C.M. and P.S.N.), the Swiss Science Foundation (PASMP3_134379/1 to J.-P.T.) and a US NIH Director's New Innovator Award (DP2OD002750 to L.A.G.). S.C.B. is supported by a Medical Scientist Training Program (MSTP) grant from the US NIH. C.E.B. is supported by a Prostate Cancer Foundation Young Investigator Award.

Author information

Authors and Affiliations

Authors

Contributions

S.C.B., M.S.L., P.S., W.M.H., E.V.A., N.S., K.C., A.S., S.L.C., G.S., D.V. and A.H.R. performed computational analyses. M.B., J.-P.T., T.A.W., S.-S.C., K.S., G.B., T.Y.M., K.P. and T.V. designed and performed experiments. E.N., D.A., R.C.O., C.G., W.W., M.C.R., K.A. and S.B.G. processed samples and supervised exome sequencing. J.M.M., K.P., N.K., A.K.T., N.R., P.J.W., H.M., C.M. and P.S.N. coordinated sample acquisition, processing, pathologic review and analysis. C.E.B., S.C.B., F.D., P.W.K., T.R.G., M.M., E.S.L., G.G., M.A.R. and L.A.G. designed the study. C.E.B., S.C.B., F.D., G.G., M.A.R. and L.A.G. analyzed the data and wrote the manuscript.

Corresponding authors

Correspondence to Mark A Rubin or Levi A Garraway.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Note, Supplementary Figures 1–18 and Supplementary Tables 1, 4–8 and 10–12 (PDF 8357 kb)

Supplementary Tables

Supplementary Tables 2, 3 and 9 (XLSX 6486 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Barbieri, C., Baca, S., Lawrence, M. et al. Exome sequencing identifies recurrent SPOP, FOXA1 and MED12 mutations in prostate cancer. Nat Genet 44, 685–689 (2012). https://doi.org/10.1038/ng.2279

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.2279

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