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:

Synthetic lethality by targeting EZH2 methyltransferase activity in ARID1A-mutated cancers

Subjects

Abstract

The gene encoding ARID1A, a chromatin remodeler, shows one of the highest mutation rates across many cancer types. Notably, ARID1A is mutated in over 50% of ovarian clear cell carcinomas, which currently have no effective therapy. To date, clinically applicable targeted cancer therapy based on ARID1A mutational status has not been described. Here we show that inhibition of the EZH2 methyltransferase acts in a synthetic lethal manner in ARID1A-mutated ovarian cancer cells and that ARID1A mutational status correlated with response to the EZH2 inhibitor. We identified PIK3IP1 as a direct target of ARID1A and EZH2 that is upregulated by EZH2 inhibition and contributed to the observed synthetic lethality by inhibiting PI3K–AKT signaling. Importantly, EZH2 inhibition caused regression of ARID1A-mutated ovarian tumors in vivo. To our knowledge, this is the first data set to demonstrate a synthetic lethality between ARID1A mutation and EZH2 inhibition. Our data indicate that pharmacological inhibition of EZH2 represents a novel treatment strategy for cancers involving ARID1A mutations.

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: GSK126, an EZH2 inhibitor, is selective against ARID1A-knockdown cells compared with controls.
Figure 2: Response to EZH2 inhibitor is dependent on ARID1A status.
Figure 3: EZH2 inhibitor triggers apoptosis of ARID1A-mutated cells.
Figure 4: PIK3IP1 is a newly recognized ARID1A–EZH2 direct target gene.
Figure 5: PIK3IP1 contributed to the observed synthetic lethality.
Figure 6: EZH2 inhibitor caused the regression and reduced the number of tumor nodules of ARID1A-mutated OCCC tumors.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Referenced accessions

Gene Expression Omnibus

References

  1. Garraway, L.A. & Lander, E.S. Lessons from the cancer genome. Cell 153, 17–37 (2013).

    Article  CAS  Google Scholar 

  2. Lawrence, M.S. et al. Discovery and saturation analysis of cancer genes across 21 tumour types. Nature 505, 495–501 (2014).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  6. Anglesio, M.S. et al. Type-specific cell line models for type-specific ovarian cancer research. PLoS ONE 8, e72162 (2013).

    Article  CAS  Google Scholar 

  7. Cao, R. & Zhang, Y. The functions of E(Z)/EZH2-mediated methylation of lysine 27 in histone H3. Curr. Opin. Genet. Dev. 14, 155–164 (2004).

    Article  CAS  Google Scholar 

  8. Li, H., Cai, Q., Godwin, A.K. & Zhang, R. Enhancer of zeste homolog 2 promotes the proliferation and invasion of epithelial ovarian cancer cells. Mol. Cancer Res. 8, 1610–1618 (2010).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  10. 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).

    Article  CAS  Google Scholar 

  11. 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).

    Article  CAS  Google Scholar 

  12. Guan, B., Gao, M., Wu, C.H., Wang, T.L. & Shih Ie, M. Functional analysis of in-frame indel ARID1A mutations reveals new regulatory mechanisms of its tumor suppressor functions. Neoplasia 14, 986–993 (2012).

    Article  CAS  Google Scholar 

  13. Yamada, K.M. & Cukierman, E. Modeling tissue morphogenesis and cancer in 3D. Cell 130, 601–610 (2007).

    Article  CAS  Google Scholar 

  14. Jenuwein, T. The epigenetic magic of histone lysine methylation. FEBS J. 273, 3121–3135 (2006).

    Article  CAS  Google Scholar 

  15. Guan, B., Wang, T.L. & Shih Ie, M. ARID1A, a factor that promotes formation of SWI/SNF-mediated chromatin remodeling, is a tumor suppressor in gynecologic cancers. Cancer Res. 71, 6718–6727 (2011).

    Article  CAS  Google Scholar 

  16. Konze, K.D. et al. An orally bioavailable chemical probe of the lysine methyltransferases EZH2 and EZH1. ACS Chem. Biol. 8, 1324–1334 (2013).

    Article  CAS  Google Scholar 

  17. Kennison, J.A. & Tamkun, J.W. Dosage-dependent modifiers of polycomb and antennapedia mutations in Drosophila. Proc. Natl. Acad. Sci. USA 85, 8136–8140 (1988).

    Article  CAS  Google Scholar 

  18. Li, H. et al. ALDH1A1 is a novel EZH2 target gene in epithelial ovarian cancer identified by genome-wide approaches. Cancer Prev. Res. (Phila.) 5, 484–491 (2012).

    Article  CAS  Google Scholar 

  19. Stany, M.P. et al. Identification of novel therapeutic targets in microdissected clear cell ovarian cancers. PLoS ONE 6, e21121 (2011).

    Article  CAS  Google Scholar 

  20. He, X. et al. PIK3IP1, a negative regulator of PI3K, suppresses the development of hepatocellular carcinoma. Cancer Res. 68, 5591–5598 (2008).

    Article  CAS  Google Scholar 

  21. Zhu, Z. et al. PI3K is negatively regulated by PIK3IP1, a novel p110 interacting protein. Biochem. Biophys. Res. Commun. 358, 66–72 (2007).

    Article  CAS  Google Scholar 

  22. Yamamoto, S., Tsuda, H., Takano, M., Tamai, S. & Matsubara, O. Loss of ARID1A protein expression occurs as an early event in ovarian clear-cell carcinoma development and frequently coexists with PIK3CA mutations. Mod. Pathol. 25, 615–624 (2012).

    Article  CAS  Google Scholar 

  23. Samartzis, E.P., Noske, A., Dedes, K.J., Fink, D. & Imesch, P. ARID1A mutations and PI3K/AKT pathway alterations in endometriosis and endometriosis-associated ovarian carcinomas. Int. J. Mol. Sci. 14, 18824–18849 (2013).

    Article  CAS  Google Scholar 

  24. Chandler, R.L. et al. ARID1a-DNA interactions are required for promoter occupancy by SWI/SNF. Mol. Cell. Biol. 33, 265–280 (2013).

    Article  CAS  Google Scholar 

  25. Davidovich, C., Zheng, L., Goodrich, K.J. & Cech, T.R. Promiscuous RNA binding by polycomb repressive complex 2. Nat. Struct. Mol. Biol. 20, 1250–1257 (2013).

    Article  CAS  Google Scholar 

  26. Cho, K.R. & Shih Ie, M. Ovarian cancer. Annu. Rev. Pathol. 4, 287–313 (2009).

    Article  CAS  Google Scholar 

  27. Helming, K.C. et al. ARID1B is a specific vulnerability in ARID1A-mutant cancers. Nat. Med. 20, 251–254 (2014).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  29. 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).

    Article  CAS  Google Scholar 

  30. Hargreaves, D.C. & Crabtree, G.R. ATP-dependent chromatin remodeling: genetics, genomics and mechanisms. Cell Res. 21, 396–420 (2011).

    Article  CAS  Google Scholar 

  31. Debnath, J., Muthuswamy, S.K. & Brugge, J.S. Morphogenesis and oncogenesis of MCF-10A mammary epithelial acini grown in three-dimensional basement membrane cultures. Methods 30, 256–268 (2003).

    Article  CAS  Google Scholar 

  32. Ye, X. et al. Downregulation of Wnt signaling is a trigger for formation of facultative heterochromatin and onset of cell senescence in primary human cells. Mol. Cell 27, 183–196 (2007).

    Article  CAS  Google Scholar 

  33. Tu, Z. et al. Oncogenic RAS regulates BRIP1 expression to induce dissociation of BRCA1 from chromatin, inhibit DNA repair, and promote senescence. Dev. Cell 21, 1077–1091 (2011).

    Article  CAS  Google Scholar 

  34. Zhang, S. A comprehensive evaluation of SAM, the SAM R-package and a simple modification to improve its performance. BMC Bioinformatics 8, 230 (2007).

    Article  Google Scholar 

  35. Storey, J.D. & Tibshirani, R. Statistical significance for genomewide studies. Proc. Natl. Acad. Sci. USA 100, 9440–9445 (2003).

    Article  CAS  Google Scholar 

  36. Langmead, B. & Salzberg, S.L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  Google Scholar 

  37. Kim, D. et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 14, R36 (2013).

    Article  Google Scholar 

  38. Bitler, B.G. et al. Wnt5a suppresses epithelial ovarian cancer by promoting cellular senescence. Cancer Res. 71, 6184–6194 (2011).

    Article  CAS  Google Scholar 

  39. Li, H. et al. SUZ12 promotes human epithelial ovarian cancer by suppressing apoptosis via silencing HRK. Mol. Cancer Res. 10, 1462–1472 (2012).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank D. Altieri, M. Murphy and R. Shiekhattar for critical comments and X. Hua and Y. Park for technical assistance. This work was supported by grants from the US National Institutes of Health/National Cancer Institute (R01CA160331 and R01CA163377 to R.Z.), a US Department of Defense Ovarian Cancer Academy award (OC093420 to R.Z.) and an Ovarian Cancer Research Fund Program project (to R.Z.). R.Z. is an Ovarian Cancer Research Fund Liz Tilberis Scholar. B.G.B. is supported by an American Cancer Society postdoctoral fellowship (PF-13-058-01-TBE). K.M.A. is supported by a training grant from the US National Institutes of Health/National Cancer Institute (T32CA9171-35). Support of Core Facilities was provided by Cancer Center Support grant CA010815 to the Wistar Institute.

Author information

Authors and Affiliations

Authors

Contributions

B.G.B. designed and performed all the experiments, analyzed data and wrote the manuscript. K.M.A. contributed to Figure 5g–i and manuscript writing. A.G. contributed to Figure 2b,c. H.L. contributed to Supplementary Figure 4g. M.A. contributed to Supplementary Figure 2c,f. A.V.K. performed the analysis presented in Figure 4a,c. D.C.S. contributed to the epigenetic-set construction. Q.L. contributed to statistical design and analysis. I.-M.S. contributed key reagents. J.R.C.-G. and D.W.S. participated in the experimental design. R.Z. conceived the study and wrote the manuscript.

Corresponding author

Correspondence to Rugang Zhang.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6 and Supplementary Tables 1–3. (PDF 13397 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bitler, B., Aird, K., Garipov, A. et al. Synthetic lethality by targeting EZH2 methyltransferase activity in ARID1A-mutated cancers. Nat Med 21, 231–238 (2015). https://doi.org/10.1038/nm.3799

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.3799

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing