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:

Discovery of a first-in-class EZH2 selective degrader

Abstract

The enhancer of zeste homolog 2 (EZH2) is the main enzymatic subunit of the PRC2 complex, which catalyzes trimethylation of histone H3 lysine 27 (H3K27me3) to promote transcriptional silencing. EZH2 is overexpressed in multiple types of cancer including triple-negative breast cancer (TNBC), and high expression levels correlate with poor prognosis. Several EZH2 inhibitors, which inhibit the methyltransferase activity of EZH2, have shown promise in treating sarcoma and follicular lymphoma in clinics. However, EZH2 inhibitors are ineffective at blocking proliferation of TNBC cells, even though they effectively reduce the H3K27me3 mark. Using a hydrophobic tagging approach, we generated MS1943, a first-in-class EZH2 selective degrader that effectively reduces EZH2 levels in cells. Importantly, MS1943 has a profound cytotoxic effect in multiple TNBC cells, while sparing normal cells, and is efficacious in vivo, suggesting that pharmacologic degradation of EZH2 can be advantageous for treating the cancers that are dependent on 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

Fig. 1: MS1943 is an EZH2 selective degrader.
Fig. 2: MS1943 (but not EZH2 inhibitors) inhibits cell growth and induces cell death in TNBC cells.
Fig. 3: Knockout or knockdown of EZH2 inhibits cell growth in TNBC cells.
Fig. 4: MS1943 suppresses tumor growth in vivo.
Fig. 5: MS1943 induces apoptosis in the MDA-MB-468 xenograft model.
Fig. 6: MS1943 results in activation of the UPR pathway.

Similar content being viewed by others

Data availability

RNA-seq data have been deposited in the GEO database (GEO accession no. GSE130503). Figure 6 and Supplementary Fig. 13 have associated raw data. There are no restrictions on data availability.

Code availability

The scripts used to analyze RNA-seq data and to produce some of the plots are available at https://github.com/parsonslabmssm/MS1943.

References

  1. Cao, R. et al. Role of histone H3 lysine 27 methylation in polycomb-group silencing. Science 298, 1039–1043 (2002).

    CAS  PubMed  Google Scholar 

  2. Kuzmichev, A., Nishioka, K., Erdjument-Bromage, H., Tempst, P. & Reinberg, D. Histone methyltransferase activity associated with a human multiprotein complex containing the enhancer of zeste protein. Genes Dev. 16, 2893–2905 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Margueron, R. & Reinberg, D. The polycomb complex PRC2 and its mark in life. Nature 469, 343–349 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Kim, K. H. & Roberts, C. W. M. Targeting EZH2 in cancer. Nat. Med. 22, 128–134 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Kaniskan, H. Ü., Martini, M. L. & Jin, J. Inhibitors of protein methyltransferases and demethylases. Chem. Rev. 118, 989–1068 (2018).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Yang, X. et al. Structure–activity relationship studies for enhancer of zeste homologue 2 (EZH2) and enhancer of zeste homologue 1 (EZH1) inhibitors. J. Med. Chem. 59, 7617–7633 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. 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  PubMed Central  Google Scholar 

  9. 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 

  10. 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 

  11. Vaswani, R. G. et al. Identification of (R)-N-((4-methoxy-6-methyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-methyl-1-(1-(1-(2,2,2-trifluoroethyl)piperidin-4-yl)ethyl)-1H-indole-3-carboxamide (CPI-1205), a potent and selective inhibitor of histone methyltransferase EZH2, suitable for phase I clinical trials for B-cell lymphomas. J. Med. Chem. 59, 9928–9941 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Kung, P. P. et al. Optimization of orally bioavailable enhancer of zeste homolog 2 (EZH2) inhibitors using ligand and property-based design strategies: identification of development candidate (R)-5,8-dichloro-7-(methoxy(oxetan-3-yl)methyl)-2-((4-methoxy-6-methyl-2-oxo-1,2- dihydropyridin-3-yl)methyl)-3,4-dihydroisoquinolin-1(2H)-one (PF-06821497). J. Med. Chem. 61, 650–665 (2018).

    CAS  PubMed  Google Scholar 

  13. 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 

  14. Zhao, Y. et al. EZH2 cooperates with gain-of-function p53 mutants to promote cancer growth and metastasis. EMBO J. 38, e99599 (2019).

    PubMed  PubMed Central  Google Scholar 

  15. Kim, J. et al. Polycomb- and methylation-independent roles of EZH2 as a transcription activator. Cell Rep. 25, 2808–2820 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Lin, N. U. et al. Clinicopathologic features, patterns of recurrence and survival among women with triple-negative breast cancer in the national comprehensive cancer network. Cancer 118, 5463–5472 (2012).

    PubMed  Google Scholar 

  17. Lara-Medina, F. et al. Triple-negative breast cancer in Hispanic patients: high prevalence, poor prognosis and association with menopausal status, body mass index and parity. Cancer 117, 3658–3669 (2011).

    PubMed  Google Scholar 

  18. Gluz, O. et al. Triple-negative breast cancer—current status and future directions. Ann. Oncol. 20, 1913–1927 (2009).

    CAS  PubMed  Google Scholar 

  19. Kleer, C. G. et al. EZH2 is a marker of aggressive breast cancer and promotes neoplastic transformation of breast epithelial cells. Proc. Natl Acad. Sci. USA 100, 11606–11611 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Varambally, S. et al. The polycomb group protein EZH2 is involved in progression of prostate cancer. Nature 419, 624–629 (2002).

    CAS  PubMed  Google Scholar 

  21. Holm, K. et al. Global H3K27 trimethylation and EZH2 abundance in breast tumor subtypes. Mol. Oncol. 6, 494–506 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Bachmann, I. M. et al. EZH2 expression is associated with high proliferation rate and aggressive tumor subgroups in cutaneous melanoma and cancers of the endometrium, prostate and breast. J. Clin. Oncol. 24, 268–273 (2006).

    CAS  PubMed  Google Scholar 

  24. Ding, L., Erdmann, C., Chinnaiyan, A. M., Merajver, S. D. & Kleer, C. G. Identification of EZH2 as a molecular marker for a precancerous state in morphologically normal breast tissues. Cancer Res. 66, 4095–4099 (2006).

    CAS  PubMed  Google Scholar 

  25. Gonzalez, M. E. et al. Downregulation of EZH2 decreases growth of estrogen receptor-negative invasive breast carcinoma and requires BRCA1. Oncogene 28, 843–853 (2009).

    PubMed  Google Scholar 

  26. Yang, X. et al. CDKN1C (p57KIP2) is a direct target of EZH2 and suppressed by multiple epigenetic mechanisms in breast cancer cells. PLoS One 4, e5011 (2009).

    PubMed  PubMed Central  Google Scholar 

  27. Chang, C. J. et al. EZH2 promotes expansion of breast tumor initiating cells through activation of RAF1-beta-catenin signaling. Cancer Cell 19, 86–100 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Curry, E. et al. Dual EZH2 and EHMT2 histone methyltransferase inhibition increases biological efficacy in breast cancer cells. Clin. Epigenetics 7, 84 (2015).

    PubMed  PubMed Central  Google Scholar 

  29. Lai, A. C. & Crews, C. M. Induced protein degradation: an emerging drug discovery paradigm. Nat. Rev. Drug Discov. 16, 101–114 (2017).

    CAS  PubMed  Google Scholar 

  30. Burslem, G. M. & Crews, C. M. Small-molecule modulation of protein homeostasis. Chem. Rev. 117, 11269–11301 (2017).

    CAS  PubMed  Google Scholar 

  31. Neklesa, T. K. & Crews, C. M. Greasy tags for protein removal. Nature 487, 308–309 (2012).

    CAS  PubMed  Google Scholar 

  32. Xie, T. et al. Pharmacological targeting of the pseudokinase Her3. Nat. Chem. Biol. 10, 1006–1012 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Fong, J. Y. et al. Therapeutic targeting of RNA splicing catalysis through inhibition of protein arginine methylation. Cancer Cell 36, 194–209 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Xu, B. et al. Selective inhibition of EZH2 and EZH1 enzymatic activity by a small molecule suppresses MLL-rearranged leukemia. Blood 125, 346–357 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Hetz, C., Chevet, E. & Harding, H. P. Targeting the unfolded protein response in disease. Nat. Rev. Drug Discov. 12, 703 (2013).

    CAS  PubMed  Google Scholar 

  38. Lee, A.-H., Iwakoshi, N. N. & Glimcher, L. H. XBP-1 regulates a subset of endoplasmic reticulum resident chaperone genes in the unfolded protein response. Mol. Cell Biol. 23, 7448–7459 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Yoshida, H., Matsui, T., Yamamoto, A., Okada, T. & Mori, K. XBP1 mRNA is induced by ATF6 and spliced by IRE1 in response to ER stress to produce a highly active transcription factor. Cell 107, 881–891 (2001).

    CAS  PubMed  Google Scholar 

  40. Cano-González, A., Mauro-Lizcano, M., Iglesias-Serret, D., Gil, J. & López-Rivas, A. Involvement of both caspase-8 and Noxa-activated pathways in endoplasmic reticulum stress-induced apoptosis in triple-negative breast tumor cells. Cell Death Dis. 9, 134 (2018).

    PubMed  PubMed Central  Google Scholar 

  41. Guo, S. et al. EZH2 overexpression in different immunophenotypes of breast carcinoma and association with clinicopathologic features. Diagn. Pathol. 11, 41 (2016).

    PubMed  PubMed Central  Google Scholar 

  42. Alford, S. H., Toy, K., Merajver, S. D. & Kleer, C. G. Increased risk for distant metastasis in patients with familial early-stage breast cancer and high EZH2 expression. Breast Cancer Res. Treat. 132, 429–437 (2012).

    CAS  PubMed  Google Scholar 

  43. Szegezdi, E., Logue, S. E., Gorman, A. M. & Samali, A. Mediators of endoplasmic reticulum stress‐induced apoptosis. EMBO Rep. 7, 880–885 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. 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 

  45. Tan, J. et al. Pharmacologic disruption of polycomb-repressive complex 2-mediated gene repression selectively induces apoptosis in cancer cells. Genes Dev. 21, 1050–1063 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Lee, J.-K. & Kim, K.-C. DZNep, inhibitor of S-adenosylhomocysteine hydrolase, down-regulates expression of SETDB1 H3K9me3 HMTase in human lung cancer cells. Biochem. Biophys. Res. Commun. 438, 647–652 (2013).

    CAS  PubMed  Google Scholar 

  47. Miranda, T. B. et al. DZNep is a global histone methylation inhibitor that reactivates developmental genes not silenced by DNA methylation. Mol. Cancer Ther. 8, 1579–1588 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Wang, X. et al. A covalently bound inhibitor triggers EZH2 degradation through ChIP‐mediated ubiquitination. EMBO J. 36, 1243–1260 (2017).

    PubMed  PubMed Central  Google Scholar 

  49. Kasibhatla, S. et al. A role for transferrin receptor in triggering apoptosis when targeted with gambogic acid. Proc. Natl Acad. Sci. USA 102, 12095–12100 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Baell, J. B. & Holloway, G. A. New substructure filters for removal of pan assay interference compounds (PAINS) from screening libraries and for their exclusion in bioassays. J. Med. Chem. 53, 2719–2740 (2010).

    CAS  PubMed  Google Scholar 

  51. Manglik, A. et al. Structure-based discovery of opioid analgesics with reduced side effects. Nature 537, 185–190 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Patro, R., Duggal, G., Love, M. I., Irizarry, R. A. & Kingsford, C. Salmon provides fast and bias-aware quantification of transcript expression. Nat. Methods 14, 417–419 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Weinstein, J. N. et al. The Cancer Genome Atlas Pan-Cancer analysis project. Nat. Genet. 45, 1113–1120 (2013).

    PubMed  PubMed Central  Google Scholar 

  54. Soneson, C., Love, M. & Robinson, M. Differential analyses for RNA-seq: transcript-level estimates improve gene-level inferences. F1000Research 4, 1521 (2016).

    PubMed Central  Google Scholar 

  55. Luo, W., Friedman, M. S., Shedden, K., Hankenson, K. D. & Woolf, P. J. GAGE: generally applicable gene set enrichment for pathway analysis. BMC Bioinformatics 10, 161 (2009).

    PubMed  PubMed Central  Google Scholar 

  56. Luo, W. & Brouwer, C. Pathview: an R/Bioconductor package for pathway-based data integration and visualization. Bioinformatics 29, 1830–1831 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Mootha, V. K. et al. PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat. Genet. 34, 267–273 (2003).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported in part by grant no. R01CA230854 (to J.J. and R.P.) from the US National Institutes of Health. J.J. also acknowledges the support by grant no. R01CA218600 from the US National Institutes of Health and an endowed professorship by the Icahn School of Medicine at Mount Sinai. B.D. acknowledges support by the Medical Scientist Training Program (MSTP) training grant no. T32GM007280 at the Icahn School of Mount Sinai from the US National Institutes of Health. We thank the National Institute of Mental Health Psychoactive Drug Screening Program (NIMH-PDSP) for generating the selectivity data of MS1943 over GPCRs, ion channels and transporters. We also thank C. Lee (Icahn School of Medicine at Mount Sinai) and W. Ma (Memorial Sloan Kettering Cancer Center) for providing reagents for shRNA knockdown experiments. This work was supported in part through the computational resources and staff expertise provided by Scientific Computing at the Icahn School of Medicine at Mount Sinai. Research reported in this paper was supported by the Office of Research Infrastructure of the US National Institutes of Health under award no. S10OD018522. The content is solely the responsibility of the authors and does not necessarily represent the official views of the US National Institutes of Health.

Author information

Authors and Affiliations

Authors

Contributions

J.J. conceived the idea. A.M., E.S., R.P. and J.J. conceived and designed the experiments. A.M. and X.Y. synthesized the compounds. K.-S.P. and A.M. performed western blot experiments. E.S. performed the proliferation and apoptosis assays, qRT–PCR, immunohistochemistry and the in vivo experiment. T.C.M. analyzed the RNA-seq, tumor weight and drug concentration data. K.-S.P., J.W. and B.D. performed cell growth inhibition assays. A.M., V.L., A.L. and S.P. performed the RNA-seq experiment. M.S., A.R. and E.G. performed EZH2 KO experiments. J.W. and E.S. performed EZH2 KD experiments. K.-S.P. and E.S. performed mechanism of action studies. All authors discussed the results and commented on the manuscript. A.M., E.S., K.-S.P., R.P. and J.J. analyzed the data and co-wrote the paper. J.W. and T.C.M. contributed equally to this work.

Corresponding authors

Correspondence to Ramon Parsons or Jian Jin.

Ethics declarations

Competing interests

J.J., R.P., A.M., E.S. and X.Y. are inventors for a patent application filed by the Icahn School of Medicine at Mount Sinai. J.J. is an equity shareholder and consultant of Cullgen.

Additional information

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

Supplementary information

Supplementary Information

Supplementary Figs. 1–22, Tables 1–3 and Supplementary Note.

Reporting Summary

Supplementary Dataset 1

Supplementary Dataset 2

Supplementary Dataset 3

Supplementary Video 1

Time-lapse pictures of MDA-MB-468 cells treated with DMSO. Cells were treated with DMSO for 3 days.

Supplementary Video 2

Time-lapse pictures of MDA-MB-468 cells treated with C24. Cells were treated with C24 (4 µM) for 3 days.

Supplementary Video 3

Time-lapse pictures of MDA-MB-468 cells treated with MS1943. Cells were treated with MS1943 (4 µM) for 3 d.

Supplementary Video 4

Time-lapse pictures of HCC1187 cells treated with DMSO. Cells were treated with DMSO for 3 d.

Supplementary Video 5

Time-lapse pictures of HCC1187 cells treated with C24. Cells were treated with C24 (4 µM) for 3 d.

Supplementary Video 6

Time-lapse pictures of HCC1187 cells treated with MS1943. Cells were treated with MS1943 (4 µM) for 3 d.

Supplementary Video 7

Time-lapse pictures of HCC70 cells treated with DMSO. Cells were treated with DMSO for 3 d.

Supplementary Video 8

Time-lapse pictures of HCC70 cells treated with C24. Cells were treated with C24 (4 µM) for 3 d.

Supplementary Video 9

Time-lapse pictures of HCC70 cells treated with MS1943. Cells were treated with MS1943 (4 µM) for 3 d.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ma, A., Stratikopoulos, E., Park, KS. et al. Discovery of a first-in-class EZH2 selective degrader. Nat Chem Biol 16, 214–222 (2020). https://doi.org/10.1038/s41589-019-0421-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-019-0421-4

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