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:

Chemoproteomic discovery of a covalent allosteric inhibitor of WRN helicase

Subjects

Abstract

WRN helicase is a promising target for treatment of cancers with microsatellite instability (MSI) due to its essential role in resolving deleterious non-canonical DNA structures that accumulate in cells with faulty mismatch repair mechanisms1,2,3,4,5. Currently there are no approved drugs directly targeting human DNA or RNA helicases, in part owing to the challenging nature of developing potent and selective compounds to this class of proteins. Here we describe the chemoproteomics-enabled discovery of a clinical-stage, covalent allosteric inhibitor of WRN, VVD-133214. This compound selectively engages a cysteine (C727) located in a region of the helicase domain subject to interdomain movement during DNA unwinding. VVD-133214 binds WRN protein cooperatively with nucleotide and stabilizes compact conformations lacking the dynamic flexibility necessary for proper helicase function, resulting in widespread double-stranded DNA breaks, nuclear swelling and cell death in MSI-high (MSI-H), but not in microsatellite-stable, cells. The compound was well tolerated in mice and led to robust tumour regression in multiple MSI-H colorectal cancer cell lines and patient-derived xenograft models. Our work shows an allosteric approach for inhibition of WRN function that circumvents competition from an endogenous ATP cofactor in cancer cells, and designates VVD-133214 as a promising drug candidate for patients with MSI-H 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

Fig. 1: Discovery of a covalent WRN inhibitor, VVD-133214, using chemoproteomics.
Fig. 2: Overall architecture of the closed conformation of the WRN helicase domain.
Fig. 3: VVD-133214 inhibits growth in MSI-H cells but not in MSS.
Fig. 4: VVD-133214 induces DNA damage-and-repair response in MSI-H cells.
Fig. 5: Pharmacokinetics/pharmacodynamics and tumour growth inhibition in MSI-H colorectal cancer xenograft models.
Fig. 6: VVD-133214 inhibits tumour growth in MSI-H PDX models.

Similar content being viewed by others

Data availability

The structural data have been deposited in wwPDB under IDs 7GQS, 7GQT and 7GQU. The mass spectrometry proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE partner repository38 with the dataset identifier PXD046214. These data can be accessed with the following username (reviewer_pxd046214@ebi.ac.uk) and password (5VPbwpt0). Proteomics analysis was performed using the Homo sapiens (2016) and Mus musculus (2017) UniProt Fasta databases. Source data are provided with this paper.

References

  1. Behan, F. M. et al. Prioritization of cancer therapeutic targets using CRISPR-Cas9 screens. Nature 568, 511–516 (2019).

    Article  ADS  CAS  PubMed  Google Scholar 

  2. Chan, E. M. et al. WRN helicase is a synthetic lethal target in microsatellite unstable cancers. Nature 568, 551–556 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  3. Kategaya, L., Perumal, S. K., Hager, J. H. & Belmont, L. D. Werner syndrome helicase is required for the survival of cancer cells with microsatellite instability. iScience 13, 488–497 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  4. Lieb, S. et al. Werner syndrome helicase is a selective vulnerability of microsatellite instability-high tumor cells. eLife 8, e43333 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  5. van Wietmarschen, N. et al. Repeat expansions confer WRN dependence in microsatellite-unstable cancers. Nature 586, 292–298 (2020).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  6. Kawakami, H., Zaanan, A. & Sinicrope, F. A. Microsatellite instability testing and its role in the management of colorectal cancer. Curr. Treat. Options Oncol. 16, 30 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Hause, R. J., Pritchard, C. C., Shendure, J. & Salipante, S. J. Classification and characterization of microsatellite instability across 18 cancer types. Nat. Med. 22, 1342–1350 (2016).

    Article  CAS  PubMed  Google Scholar 

  8. Bonneville, R. et al. Landscape of microsatellite instability across 39 cancer types. JCO Precis. Oncol. 2017, PO.17.00073 (2017).

    PubMed  Google Scholar 

  9. Andre, T. et al. Pembrolizumab in microsatellite-instability-high advanced colorectal cancer. N. Engl. J. Med. 383, 2207–2218 (2020).

    Article  CAS  PubMed  Google Scholar 

  10. Lenz, H. J. et al. First-line nivolumab plus low-dose ipilimumab for microsatellite instability-Hhigh/mismatch repair-deficient metastatic colorectal cancer: the Phase II CheckMate 142 Study. J. Clin. Oncol. 40, 161–170 (2022).

    Article  CAS  PubMed  Google Scholar 

  11. Shan, J., Han, D., Shen, C., Lei, Q. & Zhang, Y. Mechanism and strategies of immunotherapy resistance in colorectal cancer. Front. Immunol. 13, 1016646 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Wang, R. et al. Intrinsic resistance and efficacy of immunotherapy in microsatellite instability-high colorectal cancer: a systematic review and meta-analysis. Biomol. Biomed. 23, 198–208 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Fuca, G. et al. Ascites and resistance to immune checkpoint inhibition in dMMR/MSI-H metastatic colorectal and gastric cancers. J. Immunother. Cancer 10, e004001 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Sui, Q. et al. Inflammation promotes resistance to immune checkpoint inhibitors in high microsatellite instability colorectal cancer. Nat. Commun. 13, 7316 (2022).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  15. Huang, S. et al. The premature ageing syndrome protein, WRN, is a 3’->5’ exonuclease. Nat. Genet. 20, 114–116 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Gray, M. D. et al. The Werner syndrome protein is a DNA helicase. Nat. Genet. 17, 100–103 (1997).

    Article  CAS  PubMed  Google Scholar 

  17. Zong, D. et al. Comprehensive mapping of cell fates in microsatellite unstable cancer cells support dual targeting of WRN and ATR. Genes Dev. 37, 913–928 (2023).

  18. Backus, K. M. et al. Proteome-wide covalent ligand discovery in native biological systems. Nature 534, 570–574 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  19. Weerapana, E. et al. Quantitative reactivity profiling predicts functional cysteines in proteomes. Nature 468, 790–795 (2010).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  20. Parker, M. J. et al. Identification of 2-sulfonyl/sulfonamide pyrimidines as covalent inhibitors of WRN using a multiplexed high-throughput screening assay. Biochemistry 62, 2147–2160 (2023).

    Article  CAS  PubMed  Google Scholar 

  21. 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 

  22. Rudolph, M. G. & Klostermeier, D. When core competence is not enough: functional interplay of the DEAD-box helicase core with ancillary domains and auxiliary factors in RNA binding and unwinding. Biol. Chem. 396, 849–865 (2015).

    Article  CAS  PubMed  Google Scholar 

  23. Cancer Dependency Map Portal (RRID:SCR_017655). DepMap Portal https://depmap.org/portal/ (2019).

  24. Bird, J. L. et al. Recapitulation of Werner syndrome sensitivity to camptothecin by limited knockdown of the WRN helicase/exonuclease. Biogerontology 13, 49–62 (2012).

    Article  CAS  PubMed  Google Scholar 

  25. Soto-Gamez, A., Quax, W. J. & Demaria, M. Regulation of survival networks in senescent cells: from mechanisms to interventions. J. Mol. Biol. 431, 2629–2643 (2019).

    Article  CAS  PubMed  Google Scholar 

  26. Kang, K., Lee, S. B., Yoo, J. H. & Nho, C. W. Flow cytometric fluorescence pulse width analysis of etoposide-induced nuclear enlargement in HCT116 cells. Biotechnol. Lett. 32, 1045–1052 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Skog, S. & Tribukait, B. Cell size following irradiation in relation to cell cycle. Acta Radiol. Oncol. 25, 269–273 (1986).

    Article  CAS  PubMed  Google Scholar 

  28. Rogakou, E. P., Pilch, D. R., Orr, A. H., Ivanova, V. S. & Bonner, W. M. DNA double-stranded breaks induce histone H2AX phosphorylation on serine 139. J. Biol. Chem. 273, 5858–5868 (1998).

    Article  CAS  PubMed  Google Scholar 

  29. Liu, Y. et al. Patient-derived xenograft models in cancer therapy: technologies and applications. Signal Transduct. Target. Ther. 8, 160 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Overman, M. J. Overview of the management of primary colon cancer. uptodate https://www.uptodate.com/contents/overview-of-the-management-of-primary-colon-cancer (2024).

  31. Picco, G. et al. Werner helicase is a synthetic-lethal vulnerability in mismatch repair-deficient colorectal cancer refractory to targeted therapies, chemotherapy, and immunotherapy. Cancer Discov. 11, 1923–1937 (2021).

    Article  CAS  PubMed  Google Scholar 

  32. Study of HRO761 Alone or in Combination in Cancer Patients With Specific DNA Alterations Called Microsatellite Instability or Mismatch Repair Deficiency (US National Library of Medicine, 2023): https://classic.clinicaltrials.gov/show/NCT05838768.

  33. Bordas, V. et al. Triazolo-pyrimidine analogues for treating diseases connected to the inhibiton of Werner syndrome RECQ helicase (WRN). International Patent WO 2022/249060 (2022).

  34. A Study to Evaluate the Safety, Pharmacokinetics, and Anti-tumor Activity of RO7589831 in Participants with Advanced Solid Tumors (US National Library of Medicine, 2023); https://classic.clinicaltrials.gov/show/NCT06004245.

  35. Newman, J. A. et al. Crystal structure of the Bloom’s syndrome helicase indicates a role for the HRDC domain in conformational changes. Nucleic Acids Res. 43, 5221–5235 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Schwanhäusser, B. et al. Global quantification of mammalian gene expression control. Nature 473, 337–342 (2011).

  37. Sommers, J. A. et al. A high-throughput screen to identify novel small molecule inhibitors of the Werner Syndrome Helicase-Nuclease (WRN). PLoS One 14, e0210525 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Perez-Riverol, Y. et al. The PRIDE database resources in 2022: a hub for mass spectrometry-based proteomics evidences. Nucleic Acids Res. 50, D543–d552 (2022).

    Article  CAS  PubMed  Google Scholar 

  39. Diederichs, K. & Karplus, P. A. Improved R-factors for diffraction data analysis in macromolecular crystallography. Nat. Struct. Biol. 4, 269–275 (1997).

    Article  CAS  PubMed  Google Scholar 

  40. Karplus, P. A. & Diederichs, K. Linking crystallographic model and data quality. Science 336, 1030–1033 (2012).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank J. Bemis, R. Nishimura, S. Larsen and I. Stiller for programme administration; I. Mochalkin, E. Aitchison, H. Binch, D. Heer, E. Jochnowitz, A. L. Lambert, A. Rufer, R. Thoma, F. Schuler and M. Wittwer for valuable technical and scientific inputs; and B. Cravatt for helpful discussions and review of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

G.M.S., M.P.P. and T.M.K. conceptualized the project. Investigation was carried out by K.A.B., K.N.L., K.T.S., C.-C.W., M.A.H., A.N.S., X.S., T.G., S.K., J.C.G., D.C.R., B.L., M.E.R.-A., D.R.W., C.L.E., S.R., S.M.N., S.M.B., E.T., V.C., H.N.W., M.K.P., J.J.S., M.G.R., M.C., D.B. and I.C. Resources were the responsibility of P.P., C.C. and J.-M.P. K.A.B. wrote the original draft. Writing, review and editing were undertaken by J.P., P.P., J.-M.P., G.M.S., M.P.P. and T.M.K. K.A.B., S.K., J.P., A.T., J.S., L.E.B., R.T.A., D.S.W., G.M.S., M.P.P. and T.M.K. supervised the project.

Corresponding authors

Correspondence to Matthew P. Patricelli or Todd M. Kinsella.

Ethics declarations

Competing interests

K.N.L., K.T.S., M.A.H., A.N.S., X.S., T.G., S.K., J.C.G., D.C.R., B.L., M.E.R.-A., D.R.W., C.L.E., S.R., S.M.N., S.M.B., J.P., V.C., H.N.W., M.K.P., J.J.S., D.S.W., G.M.S., M.P.P. and T.M.K. are current employees of Vividion Therapeutics. K.A.B., A.T., L.E.B., R.T.A., E.T., S.R. and C.-C.W. are former employees of Vividion Therapeutics. P.P., M.G.R., M.C., D.B., C.C. and J.-M.P. are current employees of F. Hoffmann-La Roche, Ltd.

Peer review

Peer review information

Nature thanks Nicholas Larsen and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data figures and tables

Extended Data Fig. 1 Additional TE and helicase activity data.

(a) Live cell and lysate TE50s for an off-target cysteine, TNFAIP3 C54 across all profiled compounds in WRN Type I (blue) and Type II (red) inhibitor series. (b) Ratios of hWRN519–1227 helicase activity without/with 0.2 mM ATP during compound preincubation period versus ratios of TE50s in lysates/live cells show that the ATP-cooperative compounds are the same ones that show enhanced potency in live cells. (c) Global TMT cysteine profiling of VVD-127101. (d) Dose-response curves of target engagement (WRN C727) by VVD-133214 in 4 cell lines: HCT-116 (gray circles), DLD-1 (white squares), SW480 (black triangles), and OCI-AML2 (circles). The comparison between both lysate (black circles) and live cells (white circles) is demonstrated in the screening line, OCI-AML2. (e) Evaluation of VVD-133214 for inhibition of mouse WRN helicase (fragment 486–1232) (mWRN486–1232) and (f) hBLM activity with- and without ATP during compound preincubation period. (g) Lack of WRN helicase inhibition activity by a non-covalent analog of VVD-133214, VVD-129448. (h) Reaction kinetics for VVD-133214 were determined using intact protein mass spectrometry with recombinant WRN. The rate of covalent modification of WRN was observed over time with varying concentrations of inhibitor. Kobs/[I] was determined to be 4848 M−1s−1 (i) Forked DNA binding of hWRN519–1227 in presence of VVD-133214 with- and without ADP preincubation via HTRF. (j) Lack of time-dependent inhibition of WRN helicase activity by VVD-133214 after helicase reaction is initiated. (i) Evaluation of VVD-133214 on helicase activity of several WRN constructs (ATPase domain hWRN532–948, C946S, Helicase Core hWRN519–1093, Helicase Core with HRDC domain hWRN519–1227, and full length WRN hWRN1–1432). Data are presented as means ± SEM.

Extended Data Fig. 2 VVD-133214 inhibits growth in MSI-high cells.

(a) Short-term treatment of Lovo cells with 2 μM VVD-133214 followed by a washout of the compound for a 6-day assay assessing nuclear number (b) and nuclear area (c) (n = 7-8 biologically independent samples). (d) No effect on nuclear area was observed following 4-day treatment of mutant HCT-116 WRN C727A cells at any concentration tested (n = 4 biologically independent samples). Data are means +/− SEM and were analyzed by one-way ANOVA. **** p<0.0001.

Extended Data Fig. 3 Time course of p53 pathway activation, cell cycle arrest, and apoptosis by VVD-133214.

Time course of dose-response of Navitoclax (a,c) or VVD-133214 (b,d) for apoptosis as measured by Real-time Glo Annexin V assay in MSI-high HCT-116 cells (a,b) or MSS SW480 cells (c,d). DNA damage pathway activation time course assessed by Simple Western™ following 2 μM VVD-133214 treatment in MSI-High HCT-116 (e), MSI-high LoVo (f), or MSS SW480 (g). Tubulin was used as a protein loading control. Columns represent individual samples. Dose-response of VVD-133214 or etoposide at 2 μM after 48 h of treatment for flow cytometry based cell cycle assessment of 2 N and 4 N DNA content in SW48 cells (h). (i) Percentage of cells in S-phase by EdU labeling. (j) Percentage of cells in mitosis by phospho-histone H3 (Ser10) staining. (k) Gating strategies for determining DNA content, EdU incorporation and phospho-Histone H3 (Ser10). (l) Time course of cycloheximide chase in HCT-116 cells upon treatment with 1 μM VVD214. WRN degradation occurs similarly in the presence or absence of cycloheximide suggesting that it is not translationally mediated. Cyclin D1 was utilized as a marker for cycloheximide activity (half-life of approximately 30 min). (m-o) HCT-116 cells were synchronized for 12 h with 0.5 mM mimosine, washed out, treated with 2 μM VVD-133214, 10 μM etoposide or DMSO and then analyzed over time for cell cycle progression (flow cytometry) (m) and WRN protein expression (Simple Western) (n). (o) Quantitation of the Simple Western data depicted in (n). Data are presented as means ± SEM.

Extended Data Fig. 4 DNA damage and repair response in MSI-high cells.

(a) Representative images of phospho-H2AX (Ser139) in MSI-high RKO cells with VVD-133214 or etoposide for 96 h. (b-e) Quantitation of Simple Western data from Fig. 4d showing effects of VVD-133214 treatment on phospho-p53 (Ser15) (b), total p53 (c), p21 (d), and WRN (e) in HCT-116 (MSI), RKO (MSI), and SW480 (MSS) cells. (f) Time course of compound-induced loss of WRN protein in HCT-116 cells treated with VVD-133214 or Etoposide (western blot quantitation normalized to tubulin expression; n = 1). (g) Rescue of VVD-133214-induced loss of WRN protein in the presence of 10 μM Bortezomib. Time points beyond 8 h could not be accurately assessed due to induction of apoptosis at later time points when using Bortezomib in HCT-116 cells. (h) Kinetic analysis of WRN protein and mRNA levels in Nocodazole synchronized HCT-116 cells demonstrating that WRN protein and mRNA are not cell cycle regulated (n = 4, technical replicates). Micrographs are representative of two independent experiments with similar results.

Extended Data Fig. 5 Tumor growth inhibition and PD in colorectal cancer xenograft models in mice.

VVD-133214 was dosed daily at 2.5, 5, 10, or 20 mg/kg via oral administration for 4 days. HCT-116 tumor tissue was collected at 2 or 24 h after the last dose (n = 4 group) and p-p53 (Ser15) (a) and p21 (b) normalized to tubulin were quantified by Simple Western. Tumor growth was assessed in female homozygous Foxn1 <nu> mice dosed orally with 2.5, 5, 10, or 20 mg/kg VVD-133214 every day for 3 weeks and p-p53 (Ser15), p21, and WRN protein levels were measured by Simple Western with tubulin as a loading control (c). Treatment did not impact body-mass (d). Additional models (n = 4–8/group) were tested for tumor growth inhibition in mice including MSI-H LoVo (e) and SW48 (f) and MSS SW480 (g). Markers of WRN inhibition were unchanged in MSS SW480 xenografts (h). Data are means +/− SEM. Data were analyzed by one-way ANOVA.

Source Data

Extended Data Fig. 6 Characterization of PDX models.

(a) Description of PDX models. TMB = tumor mutational burden. (b) History of treatment for immunotherapy-refractory PDX model. (c-j) Spider plots of individual mice bearing the indicated PDX models treated with once-daily oral dosing of VVD-133214 at 20 mg/kg. CR = complete response.

Source Data

Extended Data Table 1 In vitro performance measures of VVD-133214
Extended Data Table 2 Data collection and refinement statistics

Supplementary information

Supplementary Figure 1

Raw data for Supplementary Fig. 1.

Reporting Summary

Source data

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Baltgalvis, K.A., Lamb, K.N., Symons, K.T. et al. Chemoproteomic discovery of a covalent allosteric inhibitor of WRN helicase. Nature (2024). https://doi.org/10.1038/s41586-024-07318-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41586-024-07318-y

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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