Skip to main content

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

  • Article
  • Published:

Sequencing of prostate cancers identifies new cancer genes, routes of progression and drug targets

Abstract

Prostate cancer represents a substantial clinical challenge because it is difficult to predict outcome and advanced disease is often fatal. We sequenced the whole genomes of 112 primary and metastatic prostate cancer samples. From joint analysis of these cancers with those from previous studies (930 cancers in total), we found evidence for 22 previously unidentified putative driver genes harboring coding mutations, as well as evidence for NEAT1 and FOXA1 acting as drivers through noncoding mutations. Through the temporal dissection of aberrations, we identified driver mutations specifically associated with steps in the progression of prostate cancer, establishing, for example, loss of CHD1 and BRCA2 as early events in cancer development of ETS fusion-negative cancers. Computational chemogenomic (canSAR) analysis of prostate cancer mutations identified 11 targets of approved drugs, 7 targets of investigational drugs, and 62 targets of compounds that may be active and should be considered candidates for future clinical trials.

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

Access options

Buy this article

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

Fig. 1: Mutational landscape of prostate cancers.
Fig. 2: Landscape of driver genes in prostate cancer.
Fig. 3: Putative new driver genes.
Fig. 4: Temporal evolution of CNAs in ETS+ and ETS prostate cancer.
Fig. 5: Heterogeneity and subclonal mutations.
Fig. 6: Clinical outcome.

Similar content being viewed by others

References

  1. Attard, G. et al. Prostate cancer. Lancet 387, 70–82 (2016).

    Article  PubMed  Google Scholar 

  2. Weischenfeldt, J. et al. Integrative genomic analyses reveal an androgen-driven somatic alteration landscape in early-onset prostate cancer. Cancer Cell 23, 159–170 (2013).

    Article  CAS  PubMed  Google Scholar 

  3. Grasso, C. S. et al. The mutational landscape of lethal castration-resistant prostate cancer. Nature 487, 239–243 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Cancer Genome Atlas Research Network. The molecular taxonomy of primary prostate cancer. Cell 163, 1011–1025 (2015).

    Article  Google Scholar 

  5. Barbieri, C. E. et al. Exome sequencing identifies recurrent SPOP, FOXA1 and MED12 mutations in prostate cancer. Nat. Genet. 44, 685–689 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Lalonde, E. et al. Tumour genomic and microenvironmental heterogeneity for integrated prediction of 5-year biochemical recurrence of prostate cancer: a retrospective cohort study. Lancet Oncol. 15, 1521–1532 (2014).

    Article  PubMed  Google Scholar 

  8. Cooper, C. S. et al. Analysis of the genetic phylogeny of multifocal prostate cancer identifies multiple independent clonal expansions in neoplastic and morphologically normal prostate tissue. Nat. Genet. 47, 367–372 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Boutros, P. C. et al. Spatial genomic heterogeneity within localized, multifocal prostate cancer. Nat. Genet. 47, 736–745 (2015).

    Article  CAS  PubMed  Google Scholar 

  10. Gundem, G. et al. The evolutionary history of lethal metastatic prostate cancer. Nature 520, 353–357 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Castro, E. et al. Effect of BRCA mutations on metastatic relapse and cause-specific survival after radical treatment for localised prostate cancer. Eur. Urol. 68, 186–193 (2015).

    Article  CAS  PubMed  Google Scholar 

  12. Kluth, M. et al. Concurrent deletion of 16q23 and PTEN is an independent prognostic feature in prostate cancer. Int. J. Cancer 137, 2354–2363 (2015).

    Article  CAS  PubMed  Google Scholar 

  13. Mosquera, J. M. et al. Concurrent AURKA and MYCN gene amplifications are harbingers of lethal treatment-related neuroendocrine prostate cancer. Neoplasia 15, 1–10 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Rodrigues, L. U. et al. Coordinate loss of MAP3K7 and CHD1 promotes aggressive prostate cancer. Cancer Res. 75, 1021–1034 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Cuzick, J. et al. Prognostic value of an RNA expression signature derived from cell cycle proliferation genes in patients with prostate cancer: a retrospective study. Lancet Oncol. 12, 245–255 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Klein, E. A. et al. Decipher genomic classifier measured on prostate biopsy predicts metastasis risk. Urology 90, 148–152 (2016).

    Article  PubMed  Google Scholar 

  17. Boström, P. J. et al. Genomic predictors of outcome in prostate cancer. Eur. Urol. 68, 1033–1044 (2015).

    Article  PubMed  Google Scholar 

  18. Luca, B.-A. et al. DESNT: a poor prognosis category of human prostate cancer. Eur. Urol. Focus. https://doi.org/10.1016/j.euf.2017.01.016 (2017).

  19. Ryan, C. J. et al. Abiraterone acetate plus prednisone versus placebo plus prednisone in chemotherapy-naive men with metastatic castration-resistant prostate cancer (COU-AA-302): final overall survival analysis of a randomised, double-blind, placebo-controlled phase 3 study. Lancet Oncol. 16, 152–160 (2015).

    Article  CAS  PubMed  Google Scholar 

  20. Loriot, Y. et al. Effect of enzalutamide on health-related quality of life, pain, and skeletal-related events in asymptomatic and minimally symptomatic, chemotherapy-naive patients with metastatic castration-resistant prostate cancer (PREVAIL): results from a randomised, phase 3 trial. Lancet Oncol. 16, 509–521 (2015).

    Article  CAS  PubMed  Google Scholar 

  21. Mateo, J. et al. DNA-repair defects and olaparib in metastatic prostate cancer. N. Engl. J. Med. 373, 1697–1708 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. James, N. D. et al. Addition of docetaxel, zoledronic acid, or both to first-line long-term hormone therapy in prostate cancer (STAMPEDE): survival results from an adaptive, multiarm, multistage, platform randomised controlled trial. Lancet 387, 1163–1177 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Forbes, S. A. et al. COSMIC: exploring the world’s knowledge of somatic mutations in human cancer. Nucleic Acids Res. 43, D805–D811 (2015).

    Article  CAS  PubMed  Google Scholar 

  24. Robinson, D. et al. Integrative clinical genomics of advanced prostate cancer. Cell 161, 1215–1228 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Baca, S. C. et al. Punctuated evolution of prostate cancer genomes. Cell 153, 666–677 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Svensson, C. et al. REST mediates androgen receptor actions on gene repression and predicts early recurrence of prostate cancer. Nucleic Acids Res. 42, 999–1015 (2014).

    Article  CAS  PubMed  Google Scholar 

  27. Liu, Z. et al. CASZ1, a candidate tumor-suppressor gene, suppresses neuroblastoma tumor growth through reprogramming gene expression. Cell Death Differ. 18, 1174–1183 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Fischer, K. & Pflugfelder, G. O. Putative breast cancer driver mutations in TBX3 cause impaired transcriptional repression. Front. Oncol. 5, 244 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  29. De Keersmaecker, K. et al. Exome sequencing identifies mutation in CNOT3 and ribosomal genes RPL5 and RPL10 in T-cell acute lymphoblastic leukemia. Nat. Genet. 45, 186–190 (2013).

    Article  PubMed  Google Scholar 

  30. Sasaki, M. et al. Regulation of the MDM2-P53 pathway and tumor growth by PICT1 via nucleolar RPL11. Nat. Med. 17, 944–951 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Chakravarty, D. et al. The oestrogen receptor alpha-regulated lncRNA NEAT1 is a critical modulator of prostate cancer. Nat. Commun. 5, 5383 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Yang, Y. A. & Yu, J. Current perspectives on FOXA1 regulation of androgen receptor signaling and prostate cancer. Genes Dis. 2, 144–151 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Takayama, K. et al. Integrative analysis of FOXP1 function reveals a tumor-suppressive effect in prostate cancer. Mol. Endocrinol. 28, 2012–2024 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Krohn, A. et al. Recurrent deletion of 3p13 targets multiple tumour suppressor genes and defines a distinct subgroup of aggressive ERG fusion-positive prostate cancers. J. Pathol. 231, 130–141 (2013).

    Article  CAS  PubMed  Google Scholar 

  35. Carver, B. S. et al. Aberrant ERG expression cooperates with loss of PTEN to promote cancer progression in the prostate. Nat. Genet. 41, 619–624 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. King, J. C. et al. Cooperativity of TMPRSS2-ERG with PI3-kinase pathway activation in prostate oncogenesis. Nat. Genet. 41, 524–526 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Kluth, M. et al. Clinical significance of different types of p53 gene alteration in surgically treated prostate cancer. Int. J. Cancer 135, 1369–1380 (2014).

    Article  CAS  PubMed  Google Scholar 

  38. Burkhardt, L. et al. CHD1 is a 5q21 tumor suppressor required for ERG rearrangement in prostate cancer. Cancer Res. 73, 2795–2805 (2013).

    Article  CAS  PubMed  Google Scholar 

  39. Liu, W. et al. Identification of novel CHD1-associated collaborative alterations of genomic structure and functional assessment of CHD1 in prostate cancer. Oncogene 31, 3939–3948 (2012).

    Article  CAS  PubMed  Google Scholar 

  40. Biankin, A. V. et al. Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature 491, 399–405 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Heun, P. SUMOrganization of the nucleus. Curr. Opin. Cell Biol. 19, 350–355 (2007).

    Article  CAS  PubMed  Google Scholar 

  42. Kaikkonen, S. et al. SUMO-specific protease 1 (SENP1) reverses the hormone-augmented SUMOylation of androgen receptor and modulates gene responses in prostate cancer cells. Mol. Endocrinol. 23, 292–307 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Smith, D. I., Zhu, Y., McAvoy, S. & Kuhn, R. Common fragile sites, extremely large genes, neural development and cancer. Cancer Lett. 232, 48–57 (2006).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Williams, J. L., Greer, P. A. & Squire, J. A. Recurrent copy number alterations in prostate cancer: an in silico meta-analysis of publicly available genomic data. Cancer Genet. 207, 474–488 (2014).

    Article  CAS  PubMed  Google Scholar 

  46. Chen, Z. et al. Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis. Nature 436, 725–730 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Bolli, N. et al. Heterogeneity of genomic evolution and mutational profiles in multiple myeloma. Nat. Commun. 5, 2997 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Alexandrov, L. B. et al. Signatures of mutational processes in human cancer. Nature 500, 415–421 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Pilati, C. et al. Mutational signature analysis identifies MUTYH deficiency in colorectal cancers and adrenocortical carcinomas. J. Pathol. 242, 10–15 (2017).

    Article  CAS  PubMed  Google Scholar 

  50. Nik-Zainal, S. et al. Landscape of somatic mutations in 560 breast cancer whole-genome sequences. Nature 534, 47–54 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Polkinghorn, W. R. et al. Androgen receptor signaling regulates DNA repair in prostate cancers. Cancer Discov. 3, 1245–1253 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Goodwin, J. F. et al. DNA-PKcs-mediated transcriptional regulation drives prostate cancer progression and metastasis. Cancer Cell 28, 97–113 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Tarish, F. L. et al. Castration radiosensitizes prostate cancer tissue by impairing DNA double-strand break repair. Sci. Transl. Med. 7, 312re11 (2015).

    Article  PubMed  Google Scholar 

  54. Tym, J. E. et al. canSAR: an updated cancer research and drug discovery knowledgebase. Nucleic Acids Res. 44, D938–D943 (2016). D1.

    Article  CAS  PubMed  Google Scholar 

  55. Leongamornlert, D. et al. Frequent germline deleterious mutations in DNA repair genes in familial prostate cancer cases are associated with advanced disease. Br. J. Cancer 110, 1663–1672 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Yang, W. et al. Genomics of Drug Sensitivity in Cancer (GDSC): a resource for therapeutic biomarker discovery in cancer cells. Nucleic Acids Res. 41, D955–D961 (2013).

    Article  CAS  PubMed  Google Scholar 

  57. Seigne, C. et al. Characterisation of prostate cancer lesions in heterozygous Men1 mutant mice. BMC Cancer 10, 395 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Malik, R. et al. Targeting the MLL complex in castration-resistant prostate cancer. Nat. Med. 21, 344–352 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Rudnicka, C. et al. Overexpression and knock-down studies highlight that a disintegrin and metalloproteinase 28 controls proliferation and migration in human prostate cancer. Medicine (Baltimore) 95, e5085 (2016).

    Article  Google Scholar 

  60. Zhang, H. et al. FOXO1 inhibits Runx2 transcriptional activity and prostate cancer cell migration and invasion. Cancer Res. 71, 3257–3267 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Malinowska, K. et al. Interleukin-6 stimulation of growth of prostate cancer in vitro and in vivo through activation of the androgen receptor. Endocr. Relat. Cancer 16, 155–169 (2009).

    Article  CAS  PubMed  Google Scholar 

  62. FitzGerald, L. M. et al. Identification of a prostate cancer susceptibility gene on chromosome 5p13q12 associated with risk of both familial and sporadic disease. Eur. J. Hum. Genet. 17, 368–377 (2009).

    Article  CAS  PubMed  Google Scholar 

  63. Zhao, W., Cao, L., Zeng, S., Qin, H. & Men, T. Upregulation of miR-556-5p promoted prostate cancer cell proliferation by suppressing PPP2R2A expression. Biomed. Pharmacother. 75, 142–147 (2015).

    Article  CAS  PubMed  Google Scholar 

  64. Parray, A. et al. ROBO1, a tumor suppressor and critical molecular barrier for localized tumor cells to acquire invasive phenotype: study in African-American and Caucasian prostate cancer models. Int. J. Cancer 135, 2493–2506 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Daniels, G. et al. TBLR1 as an androgen receptor (AR) coactivator selectively activates AR target genes to inhibit prostate cancer growth. Endocr. Relat. Cancer 21, 127–142 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Jones, S. et al. Somatic mutations in the chromatin remodeling gene ARID1A occur in several tumor types. Hum. Mutat. 33, 100–103 (2012).

    Article  CAS  PubMed  Google Scholar 

  67. Collart, M. A., Kassem, S. & Villanyi, Z. Mutations in the NOT genes or in the translation machinery similarly display increased resistance to histidine starvation. Front. Genet. 8, 61 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  68. Mao, X. et al. Distinct genomic alterations in prostate cancers in Chinese and Western populations suggest alternative pathways of prostate carcinogenesis. Cancer Res. 70, 5207–5212 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Liu, W. et al. Copy number analysis indicates monoclonal origin of lethal metastatic prostate cancer. Nat. Med. 15, 559–565 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Nickerson, M. L. et al. Somatic alterations contributing to metastasis of a castration-resistant prostate cancer. Hum. Mutat. 34, 1231–1241 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Li, H. & Durbin, R. Fast and accurate long-read alignment with Burrows-Wheeler transform. Bioinformatics 26, 589–595 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  72. Ye, K., Schulz, M. H., Long, Q., Apweiler, R. & Ning, Z. Pindel: a pattern growth approach to detect break points of large deletions and medium sized insertions from paired-end short reads. Bioinformatics 25, 2865–2871 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Nik-Zainal, S. et al. The life history of 21 breast cancers. Cell 149, 994–1007 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Nilsen, G. et al. Copynumber: efficient algorithms for single- and multi-track copy number segmentation. BMC Genomics 13, 591 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Van Loo, P. et al. Allele-specific copy number analysis of tumors. Proc. Natl. Acad Sci. USA 107, 16910–16915 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  76. Firth, D. & Turner, H. L. Bradley-Terry models in R: the BradleyTerry2 package. J. Stat. Softw. 48, 1–21 (2012).

  77. Alexandrov, L. B., Nik-Zainal, S., Wedge, D. C., Campbell, P. J. & Stratton, M. R. Deciphering signatures of mutational processes operative in human cancer. Cell Rep. 3, 246–259 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Shin, S., Fine, J. & Liu, Y. Adaptive estimation with partially overlapping models. Stat. Sin 26, 235–253 (2016).

    PubMed  PubMed Central  Google Scholar 

  79. Orchard, S. et al. Protein interaction data curation: the International Molecular Exchange (IMEx) consortium. Nat. Methods 9, 345–350 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Patel, M. N., Halling-Brown, M. D., Tym, J. E., Workman, P. & Al-Lazikani, B. Objective assessment of cancer genes for drug discovery. Nat. Rev. Drug Discov. 12, 35–50 (2013).

    Article  CAS  PubMed  Google Scholar 

  81. Bulusu, K. C., Tym, J. E., Coker, E. A., Schierz, A. C. & Al-Lazikani, B. canSAR: updated cancer research and drug discovery knowledgebase. Nucleic Acids Res. 42, D1040–D1047 (2014).

    Article  CAS  PubMed  Google Scholar 

  82. Mitsopoulos, C., Schierz, A. C., Workman, P. & Al-Lazikani, B. Distinctive behaviors of druggable proteins in cellular networks. PLoS Comput. Biol. 11, e1004597 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  83. Workman, P. & Al-Lazikani, B. Drugging cancer genomes. Nat. Rev. Drug Discov. 12, 889–890 (2013).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank those men with prostate cancer and the subjects who have donated their time and their samples to the Cambridge, Oxford, The Institute of Cancer Research, John Hopkins and University of Tampere BioMediTech Biorepositories for this study. We also acknowledge support of the research staff in S4 who so carefully curated the samples and the follow-up data (J. Burge, M. Corcoran, A. George and S. Stearn). We thank M. Stratton for discussions when setting up the CR-UK Prostate Cancer ICGC Project. We acknowledge support from Cancer Research UK C5047/A14835/A22530/A17528, C309/A11566, C368/A6743, A368/A7990, C14303/A17197 (Z.K.-J., S. Merson, N.C., S.E., D.L., T. Dadaev, M.A., E.B., J.B., G.A., P.W., B.A.-L., D.S.B., C.S.C., R.A.E.), the Dallaglio Foundation (CR-UK Prostate Cancer ICGC Project and Pan Prostate Cancer Group), PC-UK/Movember (Z.K.-J.), the NIHR support to The Biomedical Research Centre at The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust (Z.K.-J., N.D., S. Merson, N.C., S.E., D.L., T. Dadaev, S. Thomas, M.A., E.B., C.F., N.L., D.N., V.K., N.A., P.K., C.O., D.C., A.T., E.M., E.R., T. Dudderidge, S. Hazell, J.B., G.A., P.W., B.A.-L., D.S.B., C.S.C., R.A.E.), Cancer Research UK funding to The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust CRUK Centre, the National Cancer Research Institute (National Institute of Health Research (NIHR) Collaborative Study: “Prostate Cancer: Mechanisms of Progression and Treatment (PROMPT)” (grant G0500966/75466) (D.E.N., V.G.), the Li Ka Shing Foundation (D.C.W., D.J.W.) and the Academy of Finland and Cancer Society of Finland (G.S.B.). We thank the National Institute for Health Research, Hutchison Whampoa Limited, University of Cambridge and the Human Research Tissue Bank (Addenbrooke’s Hospital), which is supported by the NIHR Cambridge Biomedical Research Centre; The Core Facilities at the Cancer Research UK Cambridge Institute, Orchid and Cancer Research UK, D. Holland from the Infrastructure Management Team, and P. Clapham from the Informatics Systems Group at the Wellcome Trust Sanger Institute. D.M.B. is supported by Orchid. C.V.’s academic time was supported by the NIHR Oxford Biomedical Research Centre (Molecular Diagnostics Theme/Multimodal Pathology sub-theme). We also acknowledge support from the Bob Champion Cancer Trust, The Masonic Charitable Foundation successor to The Grand Charity, The King Family and the Stephen Hargrave Trust (C.S.C., D.S.B.). P.W. is a Cancer Research Life Fellow. We acknowledge core facilities provided by CRUK funding to the CRUK ICR Centre, the CRUK Cancer Therapeutics Unit and support for canSAR C35696/A23187 (P.W., G.A.).

Author information

Authors and Affiliations

Authors

Consortia

Contributions

R.A.E., C.S.C., D.E.N., D.S.B., C.S.F., G.S.B., A.G.L., P.W., B.A.-L., D.C.W., F.C.H. and D.F.E. designed the study. R.A.E., C.S.C., D.C.W. and D.S.B. wrote the paper. M.F., P.C.B., R.G.B. and all other authors contributed to revisions. Z.K.-J., H.W., C.E.M., D.E.N., V.G., A.G.L., R.A.E., F.C.H., G.S.B., A.Y.W., C.S.F., C.V., D.M.B., N.D., S. Merson, S. Hawkins, W.H., Y.-J.L., A.L., J.K., K.K., H.L., L. Marsden, S.E., L. Matthews, A.N., Y.Y., H.Z., S.T., E.B., C.F., N.L., S. Hazell, D.N., P.G., V.K., N.V.A., P.K., C.O., D.C., A.T., E.M., E.R., T. Dudderidge, C.C., W.I., T.V. and N.C.S. coordinated sample collection, pathology review and processing. D.C.W., G.G., T.M., I.M., D.J.W., D.S.B., M.G., J.Z., A.B., L.B.A., S.D., B.K., N.C., V.B., D.L., S. McLaren, T. Dadaev, M.A., S.T., C.G., K.R., D.J., A.M., L.S., J.T., A.F., P.V.L. and U.M. supported, directed and performed the analyses. C.S. and the TCGA, J.d.B. and G.A. provided data for the meta-analysis. D.F.E., A.G.L., G.S.B., C.S.F., D.S.B., D.E.N., C.S.C. and R.A.E. are joint principal investigators for the CR-UK Prostate Cancer ICGC Project.

Corresponding authors

Correspondence to David C. Wedge or Rosalind A. Eeles.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Integrated supplementary information

Supplementary Figure 1 RNA expression of novel driver genes.

Data are taken from the CamCaP study, which includes 40 samples from this study. All genes identified as novel within this study and bearing structural variants in at least two samples are shown. P-values are from two-sided Wilcoxon rank sum test.

Supplementary Figure 2 Heterogeneity of point substitutions and CNAs.

Each dot represents a different sample, colored by sample type. The x-axis shows the amount of each genome that has subclonal CNA divided by the total amount of each genome that is copy-number aberrant, and the y-axis shows the fraction of point substitutions that are subclonal. Contour lines were calculated using R package kde2d.

Supplementary Figure 3 Multiple driver mutations in APC.

(a) Mutations in the APC gene in PD14713a are mutually exclusive, indicating that they have occurred in separate subclonal populations. Each blue or yellow string represents a forward or backward read, with somatic variants shown in red. Mutations in the left-hand and right-hand red-boxed regions never occur on the same reads. (b) PD14713a is haploid on chromosome 5q, so the mutual exclusivity of APC mutations cannot be explained by occurrence in different chromosome copies. Purple lines show total copy number, and blue lines show minor allele specific copy number, called by the Battenberg algorithm.

Supplementary Figure 4 Prognostic biomarkers.

(a) Kaplan-Meier plot of the number of mutational processes detected vs. time to biochemical recurrence (n = 89 independent prostate cancer patients who had a prostatectomy; all primary TURP samples (3) and metastatic samples (20) were excluded from this analysis). Two-sided log rank test P value is indicated. (b) Correlation between the number of processes detected and the number of substitutions detected. There was a significant positive correlation (r = 0.31, P = 0.0027; two-sided Spearman’s correlation, n = 10, 42, 25, 10, and 2 biologically independent samples for 1, 2, 3, 4, and 5 mutational processes detected, respectively.). The number of mutational signatures identified in a cancer was negatively correlated with time to biochemical recurrence in prostatectomy patients (P = 0.014, HR = 3.0; Cox proportional hazards model on number of processes greater than three). The number of substitutions detected was also an independent prognostic biomarker (P = 0.031, HR = 1.0005; Cox proportional hazards model).

Supplementary Figure 5 Analysis pipeline to identify the prostate disease network.

Mutated proteins do not function in isolation but rather through interacting within pathways and complex networks which propagate the disease causing effect. These wider pathways and networks can provide additional avenues for therapy. To identify the prostate cancer disease network, we performed the following steps. The 71 protein products of the 73 genes identified in the study were used to seed a search in the canSAR interactome to identify direct protein-protein interactions, yielding a much larger list B. The proteins in list B were then individually expanded and only proteins enriched for interaction with the prostate disease proteins retained (see Online Methods for statistical detail). Iterative culling of the network, retaining only the most significant intreractors, finally yielded 156 proteins that are either prostate proteins or proteins that significantly interact with them. This list was then submitted to canSAR druggability and actionability analysis as described in the Online Methods.

Supplementary Figure 6 dN/dS analysis.

QQ plot of the P-values derived from dN/dS analysis shows no sign of inflation. Four genes (of 20,184) had P-values = 0 and are not shown on this plot.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6 and Supplementary Note

Reporting Summary

Supplementary Table 1

Summary of sample specific genetic aberrations. Worksheet 1 reports the number of genetic aberrations identified in each gene this study, separated by type: essential splice site, frameshift, inframe, missense, nonsense, silent, stop-lost, homozygous deletion, rearrangement (n = 112 biologically independent samples). We also report the number of samples with multiple hits resulting in homozygous loss. Worksheet 2 reports the number of genetic aberrations identified in each gene across the joint dataset (n = 930 biologically independent samples). Worksheet 3 reports the samples within our dataset that bear homozygous losses in each gene. Worksheet 4 contains results of dNdScv analysis to identify coding drivers. Novel drivers are shown with red text. Significant q-values (FDR < 0.1, Benjamini- Hochberg) from analyzing either missense SNVs or all SNVs and indels are shown with green shading. Effect sizes (dN/dS values) are reported separately for missense, nonsense, essential splice site and indel variants (n = 930 biologically independent samples). Worksheet 5 contains results of NBR analysis to identify noncoding drivers in samples within this study. Significant regions (FDR < 0.1, Benjamini-Hochberg) are shown with blue/purple shading (n = 112 biologically independent samples).

Supplementary Table 2

Classification of driver genes. Genes were identified in our study using several methods, detailed in the last column: dN/dS; enrichment for SVs or CNAs in ETS+ or ETS- cancers; enrichment for truncating mutations or homozygous deletions, clinical correlation. From a PubMed literature search, prior evidence for each gene being a driver of prostate cancer was classified as ‘low’ if the gene has not been previously reported as playing a role in prostate cancer tumorigenesis or progression. Prior evidence was classified as ‘medium’ for genes reported previously as playing a role in prostate carcinogenesis or progression but currently lacking statistical support based on genetic alterations. Evidence considered included presence of multiple genetic alterations, SNP associations, and known cancer genes in other tissues. The high confidence genes are those that are widely accepted to represent cancer genes and to be altered in prostate cancer. In each case, there are two or more of the following: statistical verification of higher incidence, biological experiments, clinical correlations, confirmation in multiple studies, recognition as cancer genes in other cancer types. In the last column, novel driver genes are classified as ‘tumor suppressor’ or ‘oncogene’ based on the predominant functional effect of the observed mutations. dN/dS, non-synonymous: synonymous ratio, calculated for all SNVs and indels; dN/dS (missense), non-synonymous: synonymous ratio calculated for missense SNVs only; SV, structural variant; CNA, copy number aberration; SNV, single nucleotide variant; indel, small insertion/deletion; ETS, E26 transformation-specific.

Supplementary Table 3

Mutational signature analysis. For each sample, the number of somatic mutations attributed to each signature is shown in worksheet 1. Worksheet 2 reports the number of mutations assigned to each signature, broken down into temporal categories (early, late, clonal, subclonal) as described in Online Methods

Supplementary Table 4

Survival analyses. Cox regression model results for the presence of recurrent in genes with time to biochemical recurrence after prostatectomy as endpoint. Multivariate analysis was performed taking into account cofactors Gleason (6–9), PSA at prostatectomy, and pathological T- stage (T2, T3). Clinical information was available for 89 prostatectomy samples with WGS data, with a median follow up of 1,108 days in which biochemical recurrence occurred in 26 patients. Red background shading indicates features that have a significant association with outcome in both univariate analyses after Benjamini-Hochberg multiple testing correction and multivariate analyses. Dark grey shading indicates features that are only significant without multiple testing correction. Light grey shading indicates features that are significant in univariate but not in multivariate analyses.

Supplementary Table 5

Druggability analysis. Genes identified using the CanSAR software. Genes are colorcoded: bright green, target of an approved drug; dark green, target of an investigational drug; yellow, target that is being investigated chemically; red, no chemical information in public databases but predicted to be druggable using our structure-based method.

Supplementary Table 6

Drug sensitivity data. Of the drugs identified through CanSAR analysis, 18 are reported in the Genomics of Drug Sensitivity in Cancer database. Of these, 5 showed significant effect on growth inhibition, and the remaining 13 drugs showed weak activity in at least one cell line.

Supplementary Table 7

Clinical and molecular details of cancers subject to DNA sequencing. Clinical characteristics of each participant are reported, including age, Gleason score, T stage, PSA and primary/metastatic. The order of samples in Fig. 1 and Fig. 2 are in the columns fig1_order and fig2_order.

Supplementary Table 8

Details of samples included and excluded from the subclonal analysis

Supplementary Table 9

Software versions used to align genomes and call variants

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wedge, D.C., Gundem, G., Mitchell, T. et al. Sequencing of prostate cancers identifies new cancer genes, routes of progression and drug targets. Nat Genet 50, 682–692 (2018). https://doi.org/10.1038/s41588-018-0086-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-018-0086-z

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research