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:

SWI/SNF-mutant cancers depend on catalytic and non-catalytic activity of EZH2

Abstract

Human cancer genome sequencing has recently revealed that genes that encode subunits of SWI/SNF chromatin remodeling complexes are frequently mutated across a wide variety of cancers, and several subunits of the complex have been shown to have bona fide tumor suppressor activity1. However, whether mutations in SWI/SNF subunits result in shared dependencies is unknown. Here we show that EZH2, a catalytic subunit of the polycomb repressive complex 2 (PRC2), is essential in all tested cancer cell lines and xenografts harboring mutations of the SWI/SNF subunits ARID1A, PBRM1, and SMARCA4, which are several of the most frequently mutated SWI/SNF subunits in human cancer, but that co-occurrence of a Ras pathway mutation is correlated with abrogation of this dependence. Notably, we demonstrate that SWI/SNF-mutant cancer cells are primarily dependent on a non-catalytic role of EZH2 in the stabilization of the PRC2 complex, and that they are only partially dependent on EZH2 histone methyltransferase activity. These results not only reveal a shared dependency of cancers with genetic alterations in SWI/SNF subunits, but also suggest that EZH2 enzymatic inhibitors now in clinical development may not fully suppress the oncogenic activity of EZH2.

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: SWI/SNF-mutant cancer cells require EZH2.
Figure 2: The catalytic activity is only partially responsible for EZH2 dependence.
Figure 3: Disruption of PRC2 stability occurs in sensitive cells after enzymatic inhibitor treatment.
Figure 4: In vivo inhibition of H3K27me3 via GSK126 caused regression of tumor growth of GSK126-sensitive cancers, but not GSK126-resistant cancers.

Similar content being viewed by others

References

  1. Kadoch, C. et al. Proteomic and bioinformatic analysis of mammalian SWI/SNF complexes identifies extensive roles in human malignancy. Nat. Genet. 45, 592–601 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Wu, J.I., Lessard, J. & Crabtree, G.R. Understanding the words of chromatin regulation. Cell 136, 200–206 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Wilson, B.G. & Roberts, C.W. SWI/SNF nucleosome remodellers and cancer. Nat. Rev. Cancer 11, 481–492 (2011).

    CAS  PubMed  Google Scholar 

  4. Weissman, B. & Knudsen, K.E. Hijacking the chromatin remodeling machinery: impact of SWI/SNF perturbations in cancer. Cancer Res. 69, 8223–8230 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Guan, B. et al. Roles of deletion of Arid1a, a tumor suppressor, in mouse ovarian tumorigenesis. J. Natl. Cancer Inst. 106, dju46 (2014).

    Google Scholar 

  6. Bultman, S.J. et al. Characterization of mammary tumors from Brg1 heterozygous mice. Oncogene 27, 460–468 (2008).

    CAS  PubMed  Google Scholar 

  7. Roberts, C.W., Leroux, M.M., Fleming, M.D. & Orkin, S.H. Highly penetrant, rapid tumorigenesis through conditional inversion of the tumor suppressor gene Snf5. Cancer Cell 2, 415–425 (2002).

    CAS  PubMed  Google Scholar 

  8. Jones, S. et al. Frequent mutations of chromatin remodeling gene ARID1A in ovarian clear cell carcinoma. Science 330, 228–231 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Wiegand, K.C. et al. ARID1A mutations in endometriosis-associated ovarian carcinomas. N. Engl. J. Med. 363, 1532–1543 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Fujimoto, A. et al. Whole-genome sequencing of liver cancers identifies etiological influences on mutation patterns and recurrent mutations in chromatin regulators. Nat. Genet. 44, 760–764 (2012).

    CAS  PubMed  Google Scholar 

  11. Li, M. et al. Inactivating mutations of the chromatin remodeling gene ARID2 in hepatocellular carcinoma. Nat. Genet. 43, 828–829 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Gui, Y. et al. Frequent mutations of chromatin remodeling genes in transitional cell carcinoma of the bladder. Nat. Genet. 43, 875–878 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Varela, I. et al. Exome sequencing identifies frequent mutation of the SWI/SNF complex gene PBRM1 in renal carcinoma. Nature 469, 539–542 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Parsons, D.W. et al. The genetic landscape of the childhood cancer medulloblastoma. Science 331, 435–439 (2011).

    CAS  PubMed  Google Scholar 

  15. Robinson, G. et al. Novel mutations target distinct subgroups of medulloblastoma. Nature 488, 43–48 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Sausen, M. et al. Integrated genomic analyses identify ARID1A and ARID1B alterations in the childhood cancer neuroblastoma. Nat. Genet. 45, 12–17 (2013).

    CAS  PubMed  Google Scholar 

  17. Shain, A.H. et al. Convergent structural alterations define SWItch/Sucrose NonFermentable (SWI/SNF) chromatin remodeler as a central tumor suppressive complex in pancreatic cancer. Proc. Natl. Acad. Sci. USA 109, E252–E259 (2012).

    CAS  PubMed  Google Scholar 

  18. Le Gallo, M. et al. Exome sequencing of serous endometrial tumors identifies recurrent somatic mutations in chromatin-remodeling and ubiquitin ligase complex genes. Nat. Genet. 44, 1310–1315 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Kennison, J.A. The Polycomb and trithorax group proteins of Drosophila: trans-regulators of homeotic gene function. Annu. Rev. Genet. 29, 289–303 (1995).

    CAS  PubMed  Google Scholar 

  20. Shao, Z. et al. Stabilization of chromatin structure by PRC1, a Polycomb complex. Cell 98, 37–46 (1999).

    CAS  PubMed  Google Scholar 

  21. Tamkun, J.W. et al. brahma: a regulator of Drosophila homeotic genes structurally related to the yeast transcriptional activator SNF2/SWI2. Cell 68, 561–572 (1992).

    CAS  PubMed  Google Scholar 

  22. Ringrose, L. & Paro, R. Epigenetic regulation of cellular memory by the Polycomb and Trithorax group proteins. Annu. Rev. Genet. 38, 413–443 (2004).

    CAS  PubMed  Google Scholar 

  23. Pasini, D., Bracken, A.P., Hansen, J.B., Capillo, M. & Helin, K. The polycomb group protein Suz12 is required for embryonic stem cell differentiation. Mol. Cell. Biol. 27, 3769–3779 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Cao, R. & Zhang, Y. SUZ12 is required for both the histone methyltransferase activity and the silencing function of the EED-EZH2 complex. Mol. Cell 15, 57–67 (2004).

    CAS  PubMed  Google Scholar 

  25. Margueron, R. et al. Role of the polycomb protein EED in the propagation of repressive histone marks. Nature 461, 762–767 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Qi, W. et al. Selective inhibition of Ezh2 by a small-molecule inhibitor blocks tumor cells proliferation. Proc. Natl. Acad. Sci. USA 109, 21360–21365 (2012).

    CAS  PubMed  Google Scholar 

  27. McCabe, M.T. et al. EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations. Nature 492, 108–112 (2012).

    CAS  PubMed  Google Scholar 

  28. McCabe, M.T. et al. Mutation of A677 in histone methyltransferase EZH2 in human B-cell lymphoma promotes hypertrimethylation of histone H3 on lysine 27 (H3K27). Proc. Natl. Acad. Sci. USA 109, 2989–2994 (2012).

    CAS  PubMed  Google Scholar 

  29. Knutson, S.K. et al. A selective inhibitor of EZH2 blocks H3K27 methylation and kills mutant lymphoma cells. Nat. Chem. Biol. 8, 890–896 (2012).

    CAS  PubMed  Google Scholar 

  30. Kia, S.K., Gorski, M.M., Giannakopoulos, S. & Verrijzer, C.P. SWI/SNF mediates polycomb eviction and epigenetic reprogramming of the INK4b-ARF-INK4a locus. Mol. Cell. Biol. 28, 3457–3464 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Knutson, S.K. et al. Durable tumor regression in genetically altered malignant rhabdoid tumors by inhibition of methyltransferase EZH2. Proc. Natl. Acad. Sci. USA 110, 7922–7927 (2013).

    CAS  PubMed  Google Scholar 

  32. Wilson, B.G. et al. Epigenetic antagonism between polycomb and SWI/SNF complexes during oncogenic transformation. Cancer Cell 18, 316–328 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Medina, P.P. et al. Frequent BRG1/SMARCA4-inactivating mutations in human lung cancer cell lines. Hum. Mutat. 29, 617–622 (2008).

    CAS  PubMed  Google Scholar 

  34. Cheung, H.W. et al. Systematic investigation of genetic vulnerabilities across cancer cell lines reveals lineage-specific dependencies in ovarian cancer. Proc. Natl. Acad. Sci. USA 108, 12372–12377 (2011).

    CAS  PubMed  Google Scholar 

  35. Shao, D.D. et al. ATARiS: computational quantification of gene suppression phenotypes from multisample RNAi screens. Genome Res. 23, 665–678 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Cowley, G.S. et al. Parallel genome-scale loss of function screens in 216 cancer cell lines for the identification of context-specific genetic dependencies. Sci. Data 1, 140035 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. MacConaill, L.E. et al. Profiling critical cancer gene mutations in clinical tumor samples. PLoS ONE 4, e7887 (2009).

    PubMed  PubMed Central  Google Scholar 

  38. De Raedt, T. et al. PRC2 loss amplifies Ras-driven transcription and confers sensitivity to BRD4-based therapies. Nature 514, 247–251 (2014).

    CAS  PubMed  Google Scholar 

  39. Kim, W. et al. Targeted disruption of the EZH2-EED complex inhibits EZH2-dependent cancer. Nat. Chem. Biol. 9, 643–650 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Wei, Y. et al. CDK1-dependent phosphorylation of EZH2 suppresses methylation of H3K27 and promotes osteogenic differentiation of human mesenchymal stem cells. Nat. Cell Biol. 13, 87–94 (2011).

    CAS  PubMed  Google Scholar 

  41. Xu, K. et al. EZH2 oncogenic activity in castration-resistant prostate cancer cells is Polycomb-independent. Science. 338, 1465–1469 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

EZ005 was provided courtesy of J. Bradner (Dana-Farber Cancer Institute). Plasmids for wild-type and SET domain–truncated (SET) EZH2 were provided courtesy of M. Brown (Dana-Farber Cancer Institute). K.H.K. was supported by an award from National Cancer Center. This work was supported by US National Institutes of Health grants R01CA172152 (C.W.M.R.), R01CA113794 (C.W.M.R.) and U01CA176058 (W.C.H.). W.K. was supported by a Claudia Adams Barr grant. T.P.H. was supported by an award from the National Institute of General Medical Sciences (T32GM007753). The Cure AT/RT Now foundation, the Avalanna Fund, the Garrett B. Smith Foundation, Miles for Mary (C.W.M.R.), a Leukemia & Lymphoma Society Specialized Center of Research Award Project Grant (L.D.W.) and the Todd J. Schwartz Memorial Fund (L.D.W.) provided additional support.

Author information

Authors and Affiliations

Authors

Contributions

K.H.K. and C.W.M.R. designed the study; K.H.K., W.W. and J.R.H. performed the experiments with the help of J.N.W.; W.K., S.H.O. and L.D.W. provided stapled peptides and contributed to data analysis; T.P.H., W.C.H., F.V. and A.T. analyzed Achilles data; K.H.K. and C.W.M.R. wrote the manuscript with comments from all authors.

Corresponding author

Correspondence to Charles W M Roberts.

Ethics declarations

Competing interests

L.D.W. is a scientific advisory board member and consultant for Aileron Therapeutics. Via the Dana-Farber Cancer Institute Novartis Drug Discovery Program, C.W.M.R. and W.C.H. receive research support and consulting fees from the Novartis Institutes for Biomedical Research.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–5 & Supplementary Table 1 (PDF 1940 kb)

Supplementary Data 1

Project Achilles data and mutation annotations (XLSX 39 kb)

Supplementary Data 2

Mutations in cell lines used in this study (XLSX 47 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kim, K., Kim, W., Howard, T. et al. SWI/SNF-mutant cancers depend on catalytic and non-catalytic activity of EZH2. Nat Med 21, 1491–1496 (2015). https://doi.org/10.1038/nm.3968

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.3968

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