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:

An allosteric PRC2 inhibitor targeting the H3K27me3 binding pocket of EED

Abstract

Polycomb repressive complex 2 (PRC2) consists of three core subunits, EZH2, EED and SUZ12, and plays pivotal roles in transcriptional regulation. The catalytic subunit EZH2 methylates histone H3 lysine 27 (H3K27), and its activity is further enhanced by the binding of EED to trimethylated H3K27 (H3K27me3). Small-molecule inhibitors that compete with the cofactor S-adenosylmethionine (SAM) have been reported. Here we report the discovery of EED226, a potent and selective PRC2 inhibitor that directly binds to the H3K27me3 binding pocket of EED. EED226 induces a conformational change upon binding EED, leading to loss of PRC2 activity. EED226 shows similar activity to SAM-competitive inhibitors in blocking H3K27 methylation of PRC2 target genes and inducing regression of human lymphoma xenograft tumors. Interestingly, EED226 also effectively inhibits PRC2 containing a mutant EZH2 protein resistant to SAM-competitive inhibitors. Together, we show that EED226 inhibits PRC2 activity via an allosteric mechanism and offers an opportunity for treatment of PRC2-dependent cancers.

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: EED226 selectively inhibits the PRC2 activity via binding to EED.
Figure 2: Crystal structure of EED–EBD–EED226 ternary complex.
Figure 3: EED226 specifically decreases cellular global and loci-specific H3K27 methylation and regulates gene expression.
Figure 4: EED226 inhibits the proliferation of DLBCL cell lines and leads to tumor regression in mouse xenograph model.
Figure 5: EED226 is effective on EZH2 inhibitor resistant mutations and synergize with EZH2 inhibitors.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Protein Data Bank

Referenced accessions

Protein Data Bank

References

  1. Kouzarides, T. Chromatin modifications and their function. Cell 128, 693–705 (2007).

    Article  CAS  PubMed  Google Scholar 

  2. Shilatifard, A. Chromatin modifications by methylation and ubiquitination: implications in the regulation of gene expression. Annu. Rev. Biochem. 75, 243–269 (2006).

    Article  CAS  PubMed  Google Scholar 

  3. Ernst, T. et al. Inactivating mutations of the histone methyltransferase gene EZH2 in myeloid disorders. Nat. Genet. 42, 722–726 (2010).

    Article  CAS  PubMed  Google Scholar 

  4. Nikoloski, G. et al. Somatic mutations of the histone methyltransferase gene EZH2 in myelodysplastic syndromes. Nat. Genet. 42, 665–667 (2010).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  6. Lee, W. et al. PRC2 is recurrently inactivated through EED or SUZ12 loss in malignant peripheral nerve sheath tumors. Nat. Genet. 46, 1227–1232 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Ntziachristos, P. et al. Genetic inactivation of the polycomb repressive complex 2 in T cell acute lymphoblastic leukemia. Nat. Med. 18, 298–301 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Bracken, A.P. et al. EZH2 is downstream of the pRB–E2F pathway, essential for proliferation and amplified in cancer. EMBO J. 22, 5323–5335 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Yu, J. et al. A polycomb repression signature in metastatic prostate cancer predicts cancer outcome. Cancer Res. 67, 10657–10663 (2007).

    Article  CAS  PubMed  Google Scholar 

  10. Wang, C. et al. EZH2 Mediates epigenetic silencing of neuroblastoma suppressor genes CASZ1, CLU, RUNX3, and NGFR. Cancer Res. 72, 315–324 (2012).

    Article  CAS  PubMed  Google Scholar 

  11. Morin, R.D. et al. Somatic mutations altering EZH2 (Tyr641) in follicular and diffuse large B-cell lymphomas of germinal-center origin. Nat. Genet. 42, 181–185 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Majer, C.R. et al. A687V EZH2 is a gain-of-function mutation found in lymphoma patients. FEBS Lett. 586, 3448–3451 (2012).

    Article  CAS  PubMed  Google Scholar 

  13. 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  PubMed Central  Google Scholar 

  14. Yap, D.B. et al. Somatic mutations at EZH2 Y641 act dominantly through a mechanism of selectively altered PRC2 catalytic activity, to increase H3K27 trimethylation. Blood 117, 2451–2459 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. 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  PubMed  PubMed Central  Google Scholar 

  16. Campbell, J.E. et al. EPZ011989, A potent, orally-available EZH2 inhibitor with robust in vivo activity. ACS Med. Chem. Lett. 6, 491–495 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Garapaty-Rao, S. et al. Identification of EZH2 and EZH1 small-molecule inhibitors with selective impact on diffuse large B cell lymphoma cell growth. Chem. Biol. 20, 1329–1339 (2013).

    Article  CAS  PubMed  Google Scholar 

  18. 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  PubMed  Google Scholar 

  19. Nasveschuk, C.G. et al. Discovery and optimization of tetramethylpiperidinyl benzamides as inhibitors of EZH2. ACS Med. Chem. Lett. 5, 378–383 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. 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  PubMed  PubMed Central  Google Scholar 

  21. 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 

  22. Brooun, A. et al. Polycomb repressive complex 2 structure with inhibitor reveals a mechanism of activation and drug resistance. Nat. Commun. 7, 11384 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. 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  PubMed  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).

    Article  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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Jiao, L. & Liu, X. Structural basis of histone H3K27 trimethylation by an active polycomb repressive complex 2. Science 350, aac4383 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Justin, N. et al. Structural basis of oncogenic histone H3K27M inhibition of human polycomb repressive complex 2. Nat. Commun. 7, 11316 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Xu, C. et al. Binding of different histone marks differentially regulates the activity and specificity of polycomb repressive complex 2 (PRC2). Proc. Natl. Acad. Sci. USA 107, 19266–19271 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Han, Z. et al. Structural basis of EZH2 recognition by EED. Structure 15, 1306–1315 (2007).

    Article  CAS  PubMed  Google Scholar 

  30. Sneeringer, C.J. et al. Coordinated activities of wild-type plus mutant EZH2 drive tumor-associated hypertrimethylation of lysine 27 on histone H3 (H3K27) in human B-cell lymphomas. Proc. Natl. Acad. Sci. USA 107, 20980–20985 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Yu, Y. et al. Quantitative profiling of combinational K27/K36 modifications on histone H3 variants in mouse organs. J. Proteome Res. 15, 1070–1079 (2016).

    Article  CAS  PubMed  Google Scholar 

  32. Yuan, W. et al. H3K36 methylation antagonizes PRC2-mediated H3K27 methylation. J. Biol. Chem. 286, 7983–7989 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Knutson, S.K. et al. Selective inhibition of EZH2 by EPZ-6438 leads to potent antitumor activity in EZH2-mutant non-Hodgkin lymphoma. Mol. Cancer Ther. 13, 842–854 (2014).

    Article  CAS  PubMed  Google Scholar 

  34. Baker, T. et al. Acquisition of a single EZH2 D1 domain mutation confers acquired resistance to EZH2-targeted inhibitors. Oncotarget 6, 32646–32655 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Gibaja, V. et al. Development of secondary mutations in wild-type and mutant EZH2 alleles cooperates to confer resistance to EZH2 inhibitors. Oncogene 35, 558–566 (2016).

    Article  CAS  PubMed  Google Scholar 

  36. Baselga, J. et al. Lapatinib with trastuzumab for HER2-positive early breast cancer (NeoALTTO): a randomised, open-label, multicentre, phase 3 trial. Lancet 379, 633–640 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Baselga, J. et al. Relationship between tumor biomarkers and efficacy in EMILIA, a phase III Study of trastuzumab emtansine in HER2-positive metastatic breast cancer. Clin. Cancer Res. 22, 3755–3763 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Mochizuki-Kashio, M. et al. Ezh2 loss in hematopoietic stem cells predisposes mice to develop heterogeneous malignancies in an Ezh1-dependent manner. Blood 126, 1172–1183 (2015).

    Article  CAS  PubMed  Google Scholar 

  39. Xu, C. & Min, J. Structure and function of WD40 domain proteins. Protein Cell 2, 202–214 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Zhang, C. & Zhang, F. The multifunctions of WD40 proteins in genome integrity and cell cycle progression. J Genomics 3, 40–50 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Stirnimann, C.U., Petsalaki, E., Russell, R.B. & Müller, C.W. WD40 proteins propel cellular networks. Trends Biochem. Sci. 35, 565–574 (2010).

    Article  CAS  PubMed  Google Scholar 

  42. Migliori, V., Mapelli, M. & Guccione, E. On WD40 proteins: propelling our knowledge of transcriptional control? Epigenetics 7, 815–822 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Cao, F. et al. Targeting MLL1 H3K4 methyltransferase activity in mixed-lineage leukemia. Mol. Cell 53, 247–261 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Li, Y. et al. Structural basis for activity regulation of MLL family methyltransferases. Nature 530, 447–452 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Antonysamy, S. et al. Crystal structure of the human PRMT5:MEP50 complex. Proc. Natl. Acad. Sci. USA 109, 17960–17965 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Ho, M.C. et al. Structure of the arginine methyltransferase PRMT5-MEP50 reveals a mechanism for substrate specificity. PLoS One 8, e57008 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Karatas, H., Townsend, E.C., Bernard, D., Dou, Y. & Wang, S. Analysis of the binding of mixed lineage leukemia 1 (MLL1) and histone 3 peptides to WD repeat domain 5 (WDR5) for the design of inhibitors of the MLL1–WDR5 interaction. J. Med. Chem. 53, 5179–5185 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Senisterra, G. et al. Small-molecule inhibition of MLL activity by disruption of its interaction with WDR5. Biochem. J. 449, 151–159 (2013).

    Article  CAS  PubMed  Google Scholar 

  49. Sackton, K.L. et al. Synergistic blockade of mitotic exit by two chemical inhibitors of the APC/C. Nature 514, 646–649 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Li, S. et al. A liquid chromatography/mass spectrometry-based generic detection method for biochemical assay and hit discovery of histone methyltransferases. Anal. Biochem. 443, 214–221 (2013).

    Article  CAS  PubMed  Google Scholar 

  51. Collaborative Computational Project, Number 4. The CCP4 suite: programs for protein crystallography. Acta Crystallogr. D Biol. Crystallogr. 50, 760–763 (1994).

  52. Emsley, P. & Cowtan, K. Coot: model-building tools for molecular graphics. Acta Crystallogr. D Biol. Crystallogr. 60, 2126–2132 (2004).

    Article  PubMed  Google Scholar 

  53. Murshudov, G.N. et al. REFMAC5 for the refinement of macromolecular crystal structures. Acta Crystallogr. D Biol. Crystallogr. 67, 355–367 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the following colleagues for their help with this manuscript: J.-H. Zhang for HTS screening; L. Zhong for cellular assay development; Y. Wei for LC–MS/MS sample analysis; M. Dillon for medicinal chemistry; L. Liu for protein expression, purification and characterization; Z. Chen for animal studies; Y. Fan for bioinformatics data handling; X. Luo and H. Liu for assistance in structural studies; Zhenting Gao for modeling analysis; Zhenhai Gao for PRC2 biology discussion; staff at SSRF for data collection.

Author information

Authors and Affiliations

Authors

Contributions

W.Q., K.Z., J.G., Y.H. and E.L. designed the study and interpreted data. K.Z., J.G., H.Z., M. Zhang, L.L., M. Zhao, Z.C., Leying F., Lijian F., Y.G., G.L., Y. Lin, M.S., Z.W., Y.Y., C. Zeng and S.Z. performed the biochemical, biophysical and structure-related experiments and data analysis. W.Q., Y.W., J.Z., L.T., S.C., C. Zhang, H.C., D.F., Q.F., H.G., X.G., Y. Liu, F.L., J.Z. and L.Z. are involved in the cellular and animal studies and data analysis. Y.H., Z.Y., A.L., L.W. and Q.Z. designed, synthesized and characterized the chemical compounds. P.A., C.O. and E.L. guided multiple aspects of this study and team collaboration. K.Z., J.G., W.Q. and E.L. wrote the manuscript with input from co-authors.

Corresponding author

Correspondence to En Li.

Ethics declarations

Competing interests

All authors performed the work herein as employees of the Novartis Institutes for Biomedical Research, Inc.

Supplementary information

Supplementary Text and Figures

Supplementary Results, Supplementary Tables 1–5 and Supplementary Figures 1–7. (PDF 1890 kb)

Supplementary Note

Synthetic Procedures. (PDF 376 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Qi, W., Zhao, K., Gu, J. et al. An allosteric PRC2 inhibitor targeting the H3K27me3 binding pocket of EED. Nat Chem Biol 13, 381–388 (2017). https://doi.org/10.1038/nchembio.2304

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nchembio.2304

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