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:

Covalent inhibition of NSD1 histone methyltransferase

Abstract

The nuclear receptor-binding SET domain (NSD) family of histone methyltransferases is associated with various malignancies, including aggressive acute leukemia with NUP98-NSD1 translocation. While NSD proteins represent attractive drug targets, their catalytic SET domains exist in autoinhibited conformation, presenting notable challenges for inhibitor development. Here, we employed a fragment-based screening strategy followed by chemical optimization, which resulted in the development of the first-in-class irreversible small-molecule inhibitors of the nuclear receptor-binding SET domain protein 1 (NSD1) SET domain. The crystal structure of NSD1 in complex with covalently bound ligand reveals a conformational change in the autoinhibitory loop of the SET domain and formation of a channel-like pocket suitable for targeting with small molecules. Our covalent lead—compound BT5—demonstrates on-target activity in NUP98-NSD1 leukemia cells, including inhibition of histone H3 lysine 36 dimethylation and downregulation of target genes, and impaired colony formation in an NUP98-NSD1 patient sample. This study will facilitate the development of the next generation of potent and selective inhibitors of the NSD histone methyltransferases.

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: Development of NSD1 ligands using a fragment-based approach.
Fig. 2: Crystal structure of NSD1 with covalently bound BT3.
Fig. 3: Characterization of the covalent engagement of irreversible NSD1 inhibitors.
Fig. 4: Characterization of the activity of NSD1 inhibitors in HMT assays.
Fig. 5: Treatment with BT5 demonstrates on-target activity in cells.
Fig. 6: Activity of BT5 in primary samples.

Similar content being viewed by others

Data availability

The crystal structure of NSD1 (PDB code no. 3OOI) was used as the search model to determine the structures reported in this study. The crystal structures of NSD1 and NSD1 in complex with BT3 have been deposited in the PDB under the accession codes 6KQP and 6KQQ, respectively. Source data are provided with this paper.

References

  1. Bennett, R. L., Swaroop, A., Troche, C. & Licht, J. D. The role of nuclear receptor-binding SET domain family histone lysine methyltransferases in cancer. Cold Spring Harb. Perspect. Med. 7, a026708 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Wagner, E. J. & Carpenter, P. B. Understanding the language of Lys36 methylation at histone H3. Nat. Rev. Mol. Cell Biol. 13, 115–126 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Morishita, M. & di Luccio, E. Cancers and the NSD family of histone lysine methyltransferases. Biochim. Biophys. Acta 1816, 158–163 (2011).

    CAS  PubMed  Google Scholar 

  4. Job, B. et al. Genomic aberrations in lung adenocarcinoma in never smokers. PLoS ONE 5, e15145 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Bianco-Miotto, T. et al. Global levels of specific histone modifications and an epigenetic gene signature predict prostate cancer progression and development. Cancer Epidemiol. Biomarkers Prev. 19, 2611–2622 (2010).

    Article  CAS  PubMed  Google Scholar 

  6. Lawrence, M. S. et al. Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature 517, 576–582 (2015).

    Article  CAS  Google Scholar 

  7. Hollink, I. H. I. M. et al. NUP98/NSD1 characterizes a novel poor prognostic group in acute myeloid leukemia with a distinct HOX gene expression pattern. Blood 118, 3645–3656 (2011).

    Article  CAS  PubMed  Google Scholar 

  8. Shiba, N. et al. NUP98-NSD1 gene fusion and its related gene expression signature are strongly associated with a poor prognosis in pediatric acute myeloid leukemia. Genes Chromosomes Cancer 52, 683–693 (2013).

    CAS  PubMed  Google Scholar 

  9. Deshpande, A. J. et al. AF10 regulates progressive H3K79 methylation and HOX gene expression in diverse AML subtypes. Cancer Cell 26, 896–908 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Wang, G. G., Cai, L., Pasillas, M. P. & Kamps, M. P. NUP98-NSD1 links H3K36 methylation to Hox-A gene activation and leukaemogenesis. Nat. Cell Biol. 9, 804–812 (2007).

    Article  CAS  PubMed  Google Scholar 

  11. Qiao, Q. et al. The structure of NSD1 reveals an autoregulatory mechanism underlying histone H3K36 methylation. J. Biol. Chem. 286, 8361–8368 (2011).

    Article  CAS  PubMed  Google Scholar 

  12. Shen, Y. et al. Identification of LEM-14 inhibitor of the oncoprotein NSD2. Biochem. Biophys. Res. Commun. 508, 102–108 (2019).

    Article  CAS  PubMed  Google Scholar 

  13. Coussens, N. P. et al. High-throughput screening with nucleosome substrate identifies small-molecule inhibitors of the human histone lysine methyltransferase NSD2. J. Biol. Chem. 293, 13750–13765 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Tisi, D. et al. Structure of the epigenetic oncogene MMSET and inhibition by N-alkyl sinefungin derivatives. ACS Chem. Biol. 11, 3093–3105 (2016).

    Article  CAS  PubMed  Google Scholar 

  15. Morrison, M. J. et al. Identification of a peptide inhibitor for the histone methyltransferase WHSC1. PLoS ONE 13, e0197082 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Hopkins, A. L., Keserü, G. M., Leeson, P. D., Rees, D. C. & Reynolds, C. H. The role of ligand efficiency metrics in drug discovery. Nat. Rev. Drug Discov. 13, 105–121 (2014).

    Article  CAS  PubMed  Google Scholar 

  17. Degani, Y., Neumann, H. & Patchornik, A. Selective cyanylation of sulfhydryl groups. J. Am. Chem. Soc. 92, 6969–6971 (1970).

    Article  CAS  PubMed  Google Scholar 

  18. Fick, R. J. et al. Sulfur-oxygen chalcogen bonding mediates AdoMet recognition in the lysine methyltransferase SET7/9. ACS Chem. Biol. 11, 748–754 (2016).

    Article  CAS  PubMed  Google Scholar 

  19. Abdeldayem, A., Raouf, Y. S., Constantinescu, S. N., Moriggl, R. & Gunning, P. T. Advances in covalent kinase inhibitors. Chem. Soc. Rev. 49, 2617–2687 (2020).

    Article  CAS  PubMed  Google Scholar 

  20. McGregor, L. M., Jenkins, M. L., Kerwin, C., Burke, J. E. & Shokat, K. M. Expanding the scope of electrophiles capable of targeting K-Ras oncogenes. Biochemistry 56, 3178–3183 (2017).

    Article  CAS  PubMed  Google Scholar 

  21. Strelow, J. M. A perspective on the kinetics of covalent and irreversible inhibition. SLAS Discov. 22, 3–20 (2017).

    Article  CAS  PubMed  Google Scholar 

  22. Rogawski, D. S. et al. Two loops undergoing concerted dynamics regulate the activity of the ASH1L histone methyltransferase. Biochemistry 54, 5401–5413 (2015).

    Article  CAS  PubMed  Google Scholar 

  23. Martinez Molina, D. et al. Monitoring drug target engagement in cells and tissues using the cellular thermal shift assay. Science 341, 84–87 (2013).

    Article  PubMed  Google Scholar 

  24. Dart, M. L. et al. Homogeneous assay for target engagement utilizing bioluminescent thermal shift. ACS Med. Chem. Lett. 9, 546–551 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Borkin, D. et al. Pharmacologic inhibition of the menin-MLL interaction blocks progression of MLL leukemia in vivo. Cancer Cell 27, 589–602 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Xu, H. et al. NUP98 fusion proteins interact with the NSL and MLL1 complexes to drive leukemogenesis. Cancer Cell 30, 863–878 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Shima, H. et al. Ring1A and Ring1B inhibit expression of Glis2 to maintain murine MOZ-TIF2 AML stem cells. Blood 131, 1833–1845 (2018).

    Article  CAS  PubMed  Google Scholar 

  28. Edmunds, J. W., Mahadevan, L. C. & Clayton, A. L. Dynamic histone H3 methylation during gene induction: HYPB/Setd2 mediates all H3K36 trimethylation. EMBO J. 27, 406–420 (2008).

    Article  CAS  PubMed  Google Scholar 

  29. Hansen, R. et al. The reactivity-driven biochemical mechanism of covalent KRASG12C inhibitors. Nat. Struct. Mol. Biol. 25, 454–462 (2018).

    Article  CAS  PubMed  Google Scholar 

  30. Eram, M. S. et al. Kinetic characterization of human histone H3 lysine 36 methyltransferases, ASH1L and SETD2. Biochim. Biophys. Acta 1850, 1842–1848 (2015).

    Article  CAS  PubMed  Google Scholar 

  31. Wu, H. et al. Structural biology of human H3K9 methyltransferases. PLoS ONE 5, e8570 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Eram, M. S. et al. Trimethylation of histone H3 lysine 36 by human methyltransferase PRDM9 protein. J. Biol. Chem. 289, 12177–12188 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Del Rizzo, P. A. et al. SET7/9 catalytic mutants reveal the role of active site water molecules in lysine multiple methylation. J. Biol. Chem. 285, 31849–31858 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Delaglio, F. et al. NMRPipe: a multidimensional spectral processing system based on UNIX pipes. J. Biomol. NMR 6, 277–293 (1995).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  36. Vagin, A. & Teplyakov, A. Molecular replacement with MOLREP. Acta Crystallogr. D Biol. Crystallogr. 66, 22–25 (2010).

    Article  CAS  PubMed  Google Scholar 

  37. Afonine, P. V. et al. Towards automated crystallographic structure refinement with phenix.refine. Acta Crystallogr. D Biol. Crystallogr. 68, 352–367 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. McCoy, A. J. et al. Phaser crystallographic software. J. Appl. Crystallogr. 40, 658–674 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Emsley, P., Lohkamp, B., Scott, W. G. & Cowtan, K. Features and development of Coot. Acta Crystallogr. D Biol. Crystallogr. 66, 486–501 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Jo, S. Y., Granowicz, E. M., Maillard, I., Thomas, D. & Hess, J. L. Requirement for Dot1l in murine postnatal hematopoiesis and leukemogenesis by MLL translocation. Blood 117, 4759–4768 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was funded by National Institute of Health (NIH) grant nos. R01 CA226759 and CA207272 to T.C., and grant nos. 1R01 CA160467 and R01 CA244254 to J.G.; Leukemia & Lymphoma Society (LLS) Translational Research Program grant nos. 6111-14 and 6564-19 to T.C., 6485-16 and 6579-20 to J.G. and LLS Scholar grant nos. 1340-17 to T.C. and 1215-14 to J.G. C.A.H. is supported by Chemistry Biology Interface Training Program (no. T32GM008597). J.G. is Rogel Scholar at the University of Michigan. This work was also supported by the Chemotherapy Foundation (A.F.) and NIH grant nos. R35 CA210065 (to A.F.) and P30 CA013696 (Flow Cytometry Shared Resource). This research used resources of the Advanced Photon Source, a U.S. Department of Energy (DOE) Office of Science User Facility operated for the DOE Office of Science by Argonne National Laboratory under contract no. DE-AC02-06CH11357. Use of the LS-CAT Sector 21 was supported by the Michigan Economic Development Corporation and the Michigan Technology Tri-Corridor (grant no. 085P1000817). The NUP98-NSD1 construct was a kind gift from A. J. Deshpande. The MOZ-TIF2 construct was provided by I. Kitabayashi and the NUP98-HOXA9 cells were provided by H. Xu.

Author information

Authors and Affiliations

Authors

Contributions

T.C. and J.G. are responsible for initiating and supervising the entire project. H.H., S.Z. and M.A.P. synthesized the compounds. H.J.C. carried out the structural biology studies. C.A.H., H.L., J.N., D.S.R., C.N., J.A. and A.H. performed the biochemical studies. S.S., K.K., P.G.A., H.M. and T.P. performed the cell biology studies. A.M., M.L.S. and A.F. provided the reagents and advised the study. All authors contributed to data analysis and the writing of the manuscript.

Corresponding authors

Correspondence to Jolanta Grembecka or Tomasz Cierpicki.

Ethics declarations

Competing interests

H.H., C.A.H., S.Z., H.J.C., M.A.P., J.G. and T.C. are coinventors on a patent application for NSD1 inhibitors. T.C. and J.G. received prior support from Kura Oncology for an unrelated project and received licensing royalties from Kura Oncology. A.F. is consulting for Ayala Pharmaceuticals and SpringWorks Therapeutics; he received previous research support from Pfizer, Bristol Myers Squibb, Merck and Eli Lilly, as well as patent and reagent licensing royalties from Novartis, EMD Millipore and Applied Biological Materials.

Additional information

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

Extended data

Extended Data Fig. 1 Crystal structure of NSD1 SET domain and mapping of the binding of BT2 using NMR.

a, Superposition of the crystal structure of NSD1 SET domain (residues 1863–2085) determined in this work (magenta) onto the previously described crystal structure of NSD1 SET (PDB code 3OOI, green). Positions of the N- and C- termini are labeled and SAM is in blue sticks. b, Ribbon (left) and surface (right) representations of NSD1 SET domain (residues 1863–2085) with mapped residues undergoing strong chemical shift perturbations (ΔσHN > 0.05ppm or ΔσN > 0.5ppm) upon binding of BT2 (shown in red).

Extended Data Fig. 2 Crystal structure of NSD1 SET domain in complex with covalently bound compound BT3.

a, The crystal structure of NSD1 SET domain (residues 1863–2085) with bound BT3 (blue), SAM (pale blue) and mapped residues undergoing strong chemical shift perturbations (ΔσHN > 0.05ppm or ΔσN > 0.5ppm) upon binding of BT3 (shown in red). Disulfide-bond dimer of BT3 found in the structure (pale green). The dimer binds in a site that is distant from inhibitor binding and lack of chemical shift perturbations in this area indicates that this represents a crystallization artifact. b, Same as in panel a rotated by 90 deg.

Extended Data Fig. 3 BT3 and BT5 bind to the same site on NSD1 SET domain.

a, b, Comparison of the two different fragments of 1H-15N HSQC spectra of 150 μM NSD1 SET domain (black), with 500 μM compound BT3 (blue) or 300 μM compound BT5 (red). Selected residues perturbed by both compounds BT3 and BT5 are boxed. c, The crystal structure of NSD1-BT3 complex showing chemical shift perturbations (ΔσHN > 0.05ppm or ΔσN > 0.5ppm) upon binding of BT3 (shown in red). Position of two cysteine residues (C2070 and C2072) involved in Zn coordination and strongly perturbed upon binding of BT3 is shown.

Extended Data Fig. 4 Compound BT5 does not bind covalently to NSD1 C2062A SET domain.

a, MS spectra of 1 μM wild-type NSD1 SET domain incubated with DMSO (left) or 16 μM BT5 (right) for 2 h showing 51% covalent engagement with BT5. b, MS spectra of 1 μM NSD1 C2062A SET domain incubated with DMSO (left) or 16 μM BT5 (right) for 2 h. No covalent engagement of BT5 is observed (expected mass 30,066 Da). Representative spectra shown are from two independent experiments (n = 2).

Extended Data Fig. 5 Treatment with BT5 lacks engagement with NSD2 and NSD3 in cells.

CETSA assay in HEK293T cells transfected with FLAG-NSD2 SET (top) or FLAG-NSD3 SET (bottom) constructs treated with DMSO or 5 μM BT5. Cell lysates were incubated for 3 min at indicated temperatures. Experiments were performed two times.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–11, Table 1 and Notes

Reporting Summary

Source data

Source Data Fig. 5

Uncropped gels for panels a, d, e, f

Source Data Extended Data Fig. 5

Uncropped gels

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Huang, H., Howard, C.A., Zari, S. et al. Covalent inhibition of NSD1 histone methyltransferase. Nat Chem Biol 16, 1403–1410 (2020). https://doi.org/10.1038/s41589-020-0626-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-020-0626-6

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