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 inhibitor of KDM5 demethylases reduces survival of drug-tolerant cancer cells

Abstract

The KDM5 family of histone demethylases catalyzes the demethylation of histone H3 on lysine 4 (H3K4) and is required for the survival of drug-tolerant persister cancer cells (DTPs). Here we report the discovery and characterization of the specific KDM5 inhibitor CPI-455. The crystal structure of KDM5A revealed the mechanism of inhibition of CPI-455 as well as the topological arrangements of protein domains that influence substrate binding. CPI-455 mediated KDM5 inhibition, elevated global levels of H3K4 trimethylation (H3K4me3) and decreased the number of DTPs in multiple cancer cell line models treated with standard chemotherapy or targeted agents. These findings show that pretreatment of cancer cells with a KDM5-specific inhibitor results in the ablation of a subpopulation of cancer cells that can serve as the founders for therapeutic relapse.

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: Identification and characterization of a potent and selective KDM5 inhibitor with reversible activity in cells.
Figure 2: CPI-455 selectively affects H3K4 methylation in several cell models.
Figure 3: Crystal structure of KDM5A.
Figure 4: Inhibitor binding at the KDM5A active site.
Figure 5: KDM5 activity is increased in DTP models in vitro and in vivo.
Figure 6: KDM5 inhibition suppresses the emergence of drug-tolerant cells.

Similar content being viewed by others

Accession codes

Primary accessions

Protein Data Bank

Referenced accessions

Protein Data Bank

References

  1. Greer, E.L. & Shi, Y. Histone methylation: a dynamic mark in health, disease and inheritance. Nat. Rev. Genet. 13, 343–357 (2012).

    Article  CAS  Google Scholar 

  2. McGrath, J. & Trojer, P. Targeting histone lysine methylation in cancer. Pharmacol. Ther. 150, 1–22 (2015).

    Article  CAS  Google Scholar 

  3. Gonzalez-Perez, A., Jene-Sanz, A. & Lopez-Bigas, N. The mutational landscape of chromatin regulatory factors across 4,623 tumor samples. Genome Biol. 14, r106 (2013).

    Article  Google Scholar 

  4. Kandoth, C. et al. Mutational landscape and significance across 12 major cancer types. Nature 502, 333–339 (2013).

    Article  CAS  Google Scholar 

  5. 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 

  6. Zack, T.I. et al. Pan-cancer patterns of somatic copy number alteration. Nat. Genet. 45, 1134–1140 (2013).

    Article  CAS  Google Scholar 

  7. Herz, H.M., Garruss, A. & Shilatifard, A. SET for life: biochemical activities and biological functions of SET domain-containing proteins. Trends Biochem. Sci. 38, 621–639 (2013).

    Article  CAS  Google Scholar 

  8. Kooistra, S.M. & Helin, K. Molecular mechanisms and potential functions of histone demethylases. Nat. Rev. Mol. Cell Biol. 13, 297–311 (2012).

    Article  CAS  Google Scholar 

  9. Upadhyay, A.K., Horton, J.R., Zhang, X. & Cheng, X. Coordinated methyl-lysine erasure: structural and functional linkage of a Jumonji demethylase domain and a reader domain. Curr. Opin. Struct. Biol. 21, 750–760 (2011).

    Article  CAS  Google Scholar 

  10. Rose, N.R., McDonough, M.A., King, O.N., Kawamura, A. & Schofield, C.J. Inhibition of 2-oxoglutarate dependent oxygenases. Chem. Soc. Rev. 40, 4364–4397 (2011).

    Article  CAS  Google Scholar 

  11. Lin, W. et al. Loss of the retinoblastoma binding protein 2 (RBP2) histone demethylase suppresses tumorigenesis in mice lacking Rb1 or Men1. Proc. Natl. Acad. Sci. USA 108, 13379–13386 (2011).

    Article  CAS  Google Scholar 

  12. Catchpole, S. et al. PLU-1/JARID1B/KDM5B is required for embryonic survival and contributes to cell proliferation in the mammary gland and in ER+ breast cancer cells. Int. J. Oncol. 38, 1267–1277 (2011).

    CAS  PubMed  Google Scholar 

  13. Yamane, K. et al. PLU-1 is an H3K4 demethylase involved in transcriptional repression and breast cancer cell proliferation. Mol. Cell 25, 801–812 (2007).

    Article  CAS  Google Scholar 

  14. Sharma, S.V. et al. A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell 141, 69–80 (2010).

    Article  CAS  Google Scholar 

  15. Roesch, A. et al. Overcoming intrinsic multidrug resistance in melanoma by blocking the mitochondrial respiratory chain of slow-cycling JARID1B(high) cells. Cancer Cell 23, 811–825 (2013).

    Article  CAS  Google Scholar 

  16. Klose, R.J., Kallin, E.M. & Zhang, Y. JmjC-domain-containing proteins and histone demethylation. Nat. Rev. Genet. 7, 715–727 (2006).

    Article  CAS  Google Scholar 

  17. Chen, Z. et al. Structural insights into histone demethylation by JMJD2 family members. Cell 125, 691–702 (2006).

    Article  CAS  Google Scholar 

  18. Dann, C.E. III, Bruick, R.K. & Deisenhofer, J. Structure of factor-inhibiting hypoxia-inducible factor 1: an asparaginyl hydroxylase involved in the hypoxic response pathway. Proc. Natl. Acad. Sci. USA 99, 15351–15356 (2002).

    Article  CAS  Google Scholar 

  19. McDonough, M.A. et al. Cellular oxygen sensing: crystal structure of hypoxia-inducible factor prolyl hydroxylase (PHD2). Proc. Natl. Acad. Sci. USA 103, 9814–9819 (2006).

    Article  CAS  Google Scholar 

  20. Rasmussen, P.B. & Staller, P. The KDM5 family of histone demethylases as targets in oncology drug discovery. Epigenomics 6, 277–286 (2014).

    Article  CAS  Google Scholar 

  21. Thinnes, C.C. et al. Targeting histone lysine demethylases—progress, challenges, and the future. Biochim. Biophys. Acta 1839, 1416–1432 (2014).

    Article  CAS  Google Scholar 

  22. Sayegh, J. et al. Identification of small molecule inhibitors of Jumonji AT-rich interactive domain 1B (JARID1B) histone demethylase by a sensitive high throughput screen. J. Biol. Chem. 288, 9408–9417 (2013).

    Article  CAS  Google Scholar 

  23. Sengoku, T. & Yokoyama, S. Structural basis for histone H3 Lys 27 demethylation by UTX/KDM6A. Genes Dev. 25, 2266–2277 (2011).

    Article  CAS  Google Scholar 

  24. Kruidenier, L. et al. A selective jumonji H3K27 demethylase inhibitor modulates the proinflammatory macrophage response. Nature 488, 404–408 (2012).

    Article  CAS  Google Scholar 

  25. Heinemann, B. et al. Inhibition of demethylases by GSK-J1/J4. Nature 514, E1–E2 (2014).

    Article  CAS  Google Scholar 

  26. Christensen, J. et al. RBP2 belongs to a family of demethylases, specific for tri-and dimethylated lysine 4 on histone 3. Cell 128, 1063–1076 (2007).

    Article  CAS  Google Scholar 

  27. Iwase, S. et al. The X-linked mental retardation gene SMCX/JARID1C defines a family of histone H3 lysine 4 demethylases. Cell 128, 1077–1088 (2007).

    Article  CAS  Google Scholar 

  28. Klose, R.J. et al. The retinoblastoma binding protein RBP2 is an H3K4 demethylase. Cell 128, 889–900 (2007).

    Article  CAS  Google Scholar 

  29. Seward, D.J. et al. Demethylation of trimethylated histone H3 Lys4 in vivo by JARID1 JmjC proteins. Nat. Struct. Mol. Biol. 14, 240–242 (2007).

    Article  CAS  Google Scholar 

  30. Ong, S.E., Mittler, G. & Mann, M. Identifying and quantifying in vivo methylation sites by heavy methyl SILAC. Nat. Methods 1, 119–126 (2004).

    Article  CAS  Google Scholar 

  31. Fodor, B.D. et al. Jmjd2b antagonizes H3K9 trimethylation at pericentric heterochromatin in mammalian cells. Genes Dev. 20, 1557–1562 (2006).

    Article  CAS  Google Scholar 

  32. Tu, S. et al. The ARID domain of the H3K4 demethylase RBP2 binds to a DNA CCGCCC motif. Nat. Struct. Mol. Biol. 15, 419–421 (2008).

    Article  CAS  Google Scholar 

  33. Iwahara, J., Iwahara, M., Daughdrill, G.W., Ford, J. & Clubb, R.T. The structure of the Dead ringer-DNA complex reveals how AT-rich interaction domains (ARIDs) recognize DNA. EMBO J. 21, 1197–1209 (2002).

    Article  CAS  Google Scholar 

  34. Roesch, A. et al. A temporarily distinct subpopulation of slow-cycling melanoma cells is required for continuous tumor growth. Cell 141, 583–594 (2010).

    Article  CAS  Google Scholar 

  35. Raha, D. et al. The cancer stem cell marker aldehyde dehydrogenase is required to maintain a drug-tolerant tumor cell subpopulation. Cancer Res. 74, 3579–3590 (2014).

    Article  CAS  Google Scholar 

  36. Horton, J.R. et al. Enzymatic and structural insights for substrate specificity of a family of jumonji histone lysine demethylases. Nat. Struct. Mol. Biol. 17, 38–43 (2010).

    Article  CAS  Google Scholar 

  37. 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  Google Scholar 

  38. Maile, T.M. et al. Mass spectrometric quantification of histone posttranslational modifications by a hybrid chemical labeling method. Mol. Cell. Proteomics 14, 1148–1158 (2015).

    Article  CAS  Google Scholar 

  39. MacLean, B. et al. Skyline: an open source document editor for creating and analyzing targeted proteomics experiments. Bioinformatics 26, 966–968 (2010).

    Article  CAS  Google Scholar 

  40. Garcia, B.A. et al. Chemical derivatization of histones for facilitated analysis by mass spectrometry. Nat. Protoc. 2, 933–938 (2007).

    Article  CAS  Google Scholar 

  41. Olsen, J.V. et al. Parts per million mass accuracy on an Orbitrap mass spectrometer via lock mass injection into a C-trap. Mol. Cell. Proteomics 4, 2010–2021 (2005).

    Article  CAS  Google Scholar 

  42. Bradley, W.D. et al. EZH2 inhibitor efficacy in non-Hodgkin's lymphoma does not require suppression of H3K27 monomethylation. Chem. Biol. 21, 1463–1475 (2014).

    Article  CAS  Google Scholar 

  43. Otwinowski, Z. & Minor, W. Processing of X-ray diffraction data collected in oscillation mode. Methods Enzymol. 276, 307–326 (1997).

    Article  CAS  Google Scholar 

  44. Adams, P.D. et al. PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta Crystallogr. D Biol. Crystallogr. 66, 213–221 (2010).

    Article  CAS  Google Scholar 

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

    Article  Google Scholar 

Download references

Acknowledgements

We thank the Genentech Structural Biology Expression Group and J. Wu for additional protein expression and purification, as well as other members of the KDM5 team. We also thank J. Settleman for comments on the manuscript and B. Haley for short hairpin design. We thank Shamrock Structures, LLC, for diffraction data collection from beamline 08ID-1 at the Canadian Light Source, supported by the Natural Sciences and Engineering Research Council of Canada, the National Research Council Canada, the Canadian Institutes of Health Research, the Province of Saskatchewan, Western Economic Diversification Canada, and the University of Saskatchewan, and from beamline 5.0.2 of the Advanced Light Source. The Berkeley Center for Structural Biology is supported in part by the National Institutes of Health, National Institute of General Medical Sciences, and the Howard Hughes Medical Institute. The Advanced Light Source is supported by the Director, Office of Science and Office of Basic Energy Sciences, of the US Department of Energy under Contract No. DE-AC02-05CH11231.

Author information

Authors and Affiliations

Authors

Contributions

M.V. and J.R.K. were involved in the crystallography efforts. V.S.G., A.G., K.E.W., P.G., W.M., E.M.F., F.L., S.O., A.G.C., Y.L., C.B., S.B., R.T.C., B.K.A. and J.-C.H. were involved in assay development, protein production, biochemical assays, and the development and execution of cell-based assays. F.L. made the KDM5A constructs. S.A., C.A.T., C.W., G.D.G., H.K., T.L., M.C. and R.P. were involved in the design and execution of DTP and ALDH assays. M.W., Y.Y., E.J. and G.V.H. were involved in the in vivo analysis of tumors in patients and mice. T.K.C., T.M.M., D.A. and J.B. participated in histone MS analysis. J.R.K., P.T. and M.C. contributed equally, oversaw all experiments and wrote the manuscript.

Corresponding authors

Correspondence to James R Kiefer, Patrick Trojer or Marie Classon.

Ethics declarations

Competing interests

V.S.G., S.A., K.E.W., W.M., J.B., S.O., C.B., S.B., R.T.C. and P.T. are employees and shareholders of Constellation Pharmaceutical Inc. P.G., F.L., J.-C.H. and B.K.A. are shareholders of Constellation Pharmaceutical Inc. All other authors are employees of Genentech Inc. and may hold stock in the Roche Group.

Supplementary information

Supplementary Text and Figures

Supplementary Results, Supplementary Tables 1–9 and Supplementary Figures 1–3. (PDF 2866 kb)

Supplementary Note

Synthetic Procedures (PDF 578 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Vinogradova, M., Gehling, V., Gustafson, A. et al. An inhibitor of KDM5 demethylases reduces survival of drug-tolerant cancer cells. Nat Chem Biol 12, 531–538 (2016). https://doi.org/10.1038/nchembio.2085

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nchembio.2085

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