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:

Sulfopin is a covalent inhibitor of Pin1 that blocks Myc-driven tumors in vivo

Abstract

The peptidyl-prolyl isomerase, Pin1, is exploited in cancer to activate oncogenes and inactivate tumor suppressors. However, despite considerable efforts, Pin1 has remained an elusive drug target. Here, we screened an electrophilic fragment library to identify covalent inhibitors targeting Pin1’s active site Cys113, leading to the development of Sulfopin, a nanomolar Pin1 inhibitor. Sulfopin is highly selective, as validated by two independent chemoproteomics methods, achieves potent cellular and in vivo target engagement and phenocopies Pin1 genetic knockout. Pin1 inhibition had only a modest effect on cancer cell line viability. Nevertheless, Sulfopin induced downregulation of c-Myc target genes, reduced tumor progression and conferred survival benefit in murine and zebrafish models of MYCN-driven neuroblastoma, and in a murine model of pancreatic cancer. Our results demonstrate that Sulfopin is a chemical probe suitable for assessment of Pin1-dependent pharmacology in cells and in vivo, and that Pin1 warrants further investigation as a potential cancer drug target.

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 Pin1-binding fragment.
Fig. 2: Sulfopin engages Pin1 in cells and in vivo.
Fig. 3: Sulfopin is highly selective for Pin1 C113 in cells.
Fig. 4: Sulfopin shows a Pin1-dependent viability effect following long-term exposure.
Fig. 5: Sulfopin downregulates Myc transcription.
Fig. 6: Sulfopin abrogates neuroblastoma and PDAC growth in vivo.

Similar content being viewed by others

Data availability

Covalent fragment screen data are provided as an Excel file (Supplementary Dataset 1). CITe-Id and rdTOP proteomics data are provided as an Excel file (Supplementary Dataset 3). RNA-seq differential expression data analysis and Enrichr results are provided as an excel file (Supplementary Datasets 5 and 6) and have been deposited with GEO (GSE166786) All structural data have been deposited in the PDB (PDB code 6VAJ). Source data are provided with this paper.

References

  1. Pawson, T. & Scott, J. D. Protein phosphorylation in signaling—50 years and counting. Trends Biochem. Sci. 30, 286–290 (2005).

    Article  CAS  PubMed  Google Scholar 

  2. Lu, K. P. & Zhou, X. Z. The prolyl isomerase PIN1: a pivotal new twist in phosphorylation signalling and disease. Nat. Rev. Mol. Cell Biol. 8, 904–916 (2007).

    Article  CAS  PubMed  Google Scholar 

  3. Lam, P. B. et al. Prolyl isomerase Pin1 is highly expressed in Her2-positive breast cancer and regulates erbB2 protein stability. Mol. Cancer 7, 91 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Shen, M., Stukenberg, P. T., Kirschner, M. W. & Lu, K. P. The essential mitotic peptidyl-prolyl isomerase Pin1 binds and regulates mitosis-specific phosphoproteins. Genes Dev. 12, 706–720 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Ryo, A., Nakamura, M., Wulf, G., Liou, Y. C. & Lu, K. P. Pin1 regulates turnover and subcellular localization of beta-catenin by inhibiting its interaction with APC. Nat. Cell Biol. 3, 793–801 (2001).

    Article  CAS  PubMed  Google Scholar 

  6. Zhou, X. Z. et al. Pin1-dependent prolyl isomerization regulates dephosphorylation of Cdc25C and tau proteins. Mol. Cell 6, 873–883 (2000).

    Article  CAS  PubMed  Google Scholar 

  7. Bao, L. et al. Prevalent overexpression of prolyl isomerase Pin1 in human cancers. Am. J. Pathol. 164, 1727–1737 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Tan, X. et al. Pin1 expression contributes to lung cancer: prognosis and carcinogenesis. Cancer Biol. Ther. 9, 111–119 (2010).

    Article  CAS  PubMed  Google Scholar 

  9. Li, Q. et al. The rs2233678 polymorphism in PIN1 promoter region reduced cancer risk: a meta-analysis. PLoS ONE 8, e68148 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Chen, Y. et al. Prolyl isomerase Pin1: a promoter of cancer and a target for therapy. Cell Death Dis. 9, 883 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Ryo, A. et al. Regulation of NF-kappaB signaling by Pin1-dependent prolyl isomerization and ubiquitin-mediated proteolysis of p65/RelA. Mol. Cell 12, 1413–1426 (2003).

    Article  CAS  PubMed  Google Scholar 

  12. Farrell, A. S. et al. Pin1 regulates the dynamics of c-Myc DNA binding to facilitate target gene regulation and oncogenesis. Mol. Cell. Biol. 33, 2930–2949 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Girardini, J. E. et al. A Pin1/mutant p53 axis promotes aggressiveness in breast cancer. Cancer Cell 20, 79–91 (2011).

    Article  CAS  PubMed  Google Scholar 

  14. Wulf, G., Garg, P., Liou, Y.-C., Iglehart, D. & Lu, K. P. Modeling breast cancer in vivo and ex vivo reveals an essential role of Pin1 in tumorigenesis. EMBO J. 23, 3397–3407 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. D’Artista, L. et al. Pin1 is required for sustained B cell proliferation upon oncogenic activation of Myc. Oncotarget 7, 21786–21798 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Boldetti, A., Guarnaccia, V., Rambaldi, A. & Parrella, E. Inhibition of the peptidyl-prolyl-isomerase Pin1 enhances the responses of acute myeloid leukemia cells to retinoic acid via stabilization of RARα and PML-RARα. Cancer Res. 69, 1016–1026 (2009).

    Article  PubMed  CAS  Google Scholar 

  17. Liu, P. H. et al. An IRAK1-PIN1 signalling axis drives intrinsic tumour resistance to radiation therapy. Nat. Cell Biol. 21, 203–213 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Rustighi, A. et al. Prolyl-isomerase Pin1 controls normal and cancer stem cells of the breast. EMBO Mol. Med. 6, 99–119 (2014).

    Article  CAS  PubMed  Google Scholar 

  19. Tsherniak, A. et al. Defining a cancer dependency map. Cell 170, 564–576 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Lee, T. H. et al. Essential role of Pin1 in the regulation of TRF1 stability and telomere maintenance. Nat. Cell Biol. 11, 97–105 (2009).

    Article  CAS  PubMed  Google Scholar 

  21. Zhou, X. Z. & Lu, K. P. The isomerase PIN1 controls numerous cancer-driving pathways and is a unique drug target. Nat. Rev. Cancer 16, 463–478 (2016).

    Article  CAS  PubMed  Google Scholar 

  22. Hennig, L. et al. Selective inactivation of parvulin-like peptidyl-prolyl cis/trans isomerases by juglone. Biochemistry 37, 5953–5960 (1998).

    Article  CAS  PubMed  Google Scholar 

  23. Wei, S. et al. Active Pin1 is a key target of all-trans retinoic acid in acute promyelocytic leukemia and breast cancer. Nat. Med. 21, 457–466 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Kozono, S. et al. Arsenic targets Pin1 and cooperates with retinoic acid to inhibit cancer-driving pathways and tumor-initiating cells. Nat. Commun. 9, 3069 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Campaner, E. et al. A covalent PIN1 inhibitor selectively targets cancer cells by a dual mechanism of action. Nat. Commun. 8, 15772 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Moore, J. D. & Potter, A. Pin1 inhibitors: pitfalls, progress and cellular pharmacology. Bioorg. Med. Chem. Lett. 23, 4283–4291 (2013).

    Article  CAS  PubMed  Google Scholar 

  27. Pinch, B. J. et al. Identification of a potent and selective covalent Pin1 inhibitor. Nat. Chem. Biol. 16, 979–987 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Resnick, E. et al. Rapid covalent-probe discovery by electrophile-fragment screening. J. Am. Chem. Soc. 141, 8951–8968 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. London, N. et al. Covalent docking of large libraries for the discovery of chemical probes. Nat. Chem. Biol. 10, 1066–1072 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Zhang, Y. et al. Structural basis for high-affinity peptide inhibition of human Pin1. ACS Chem. Biol. 2, 320–328 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Yaffe, M. B. et al. Sequence-specific and phosphorylation-dependent proline isomerization: a potential mitotic regulatory mechanism. Science 278, 1957–1960 (1997).

    Article  CAS  PubMed  Google Scholar 

  32. Guo, C. et al. Structure-based design of novel human Pin1 inhibitors (III): optimizing affinity beyond the phosphate recognition pocket. Bioorg. Med. Chem. Lett. 24, 4187–4191 (2014).

    Article  CAS  PubMed  Google Scholar 

  33. Browne, C. M. et al. A chemoproteomic strategy for direct and proteome-wide covalent inhibitor target-site identification. J. Am. Chem. Soc. 141, 191–203 (2019).

    Article  CAS  PubMed  Google Scholar 

  34. Yang, F., Gao, J., Che, J., Jia, G. & Wang, C. A dimethyl-labeling-based strategy for site-specifically quantitative chemical proteomics. Anal. Chem. 90, 9576–9582 (2018).

    Article  CAS  PubMed  Google Scholar 

  35. Phan, R. T., Saito, M., Kitagawa, Y., Means, A. R. & Dalla-Favera, R. Genotoxic stress regulates expression of the proto-oncogene Bcl6 in germinal center B cells. Nat. Immunol. 8, 1132–1139 (2007).

    Article  CAS  PubMed  Google Scholar 

  36. Yu, C. et al. High-throughput identification of genotype-specific cancer vulnerabilities in mixtures of barcoded tumor cell lines. Nat. Biotechnol. 34, 419–423 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Fila, C., Metz, C. & van der Sluijs, P. Juglone inactivates cysteine-rich proteins required for progression through mitosis. J. Biol. Chem. 283, 21714–21724 (2008).

    Article  CAS  PubMed  Google Scholar 

  38. Pelengaris, S., Khan, M. & Evan, G. c-MYC: more than just a matter of life and death. Nat. Rev. Cancer 2, 764–776 (2002).

    Article  CAS  PubMed  Google Scholar 

  39. Yeh, E. et al. A signalling pathway controlling c-Myc degradation that impacts oncogenic transformation of human cells. Nat. Cell Biol. 6, 308–318 (2004).

    Article  CAS  PubMed  Google Scholar 

  40. Su, Y. et al. Post-translational modification localizes MYC to the nuclear pore basket to regulate a subset of target genes involved in cellular responses to environmental signals. Genes Dev. 32, 1398–1419 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Kuleshov, M. V. et al. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 44, W90–W97 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. He, S. et al. Synergy between loss of NF1 and overexpression of MYCN in neuroblastoma is mediated by the GAP-related domain. eLife 5, e14713 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. He, S. et al. Neutrophil-mediated experimental metastasis is enhanced by VEGFR inhibition in a zebrafish xenograft model. J. Pathol. 227, 431–445 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Chesler, L. & Weiss, W. A. Genetically engineered murine models – contribution to our understanding of the genetics, molecular pathology and therapeutic targeting of neuroblastoma. Semin. Cancer Biol. 21, 245–255 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Chao, S. H., Greenleaf, A. L. & Price, D. H. Juglone, an inhibitor of the peptidyl-prolyl isomerase Pin1, also directly blocks transcription. Nucleic Acids Res. 29, 767–773 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Dong, L. et al. Structure-based design of novel human Pin1 inhibitors (II). Bioorg. Med. Chem. Lett. 20, 2210–2214 (2010).

    Article  CAS  PubMed  Google Scholar 

  47. Guo, C. et al. Structure-based design of novel human Pin1 inhibitors (I). Bioorg. Med. Chem. Lett. 19, 5613–5616 (2009).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Salzberg, Y. et al. Synaptic protein degradation controls sexually dimorphic circuits through regulation of DCC/UNC-40. Curr. Biol. 30, 4128–4141 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Janes, M. R. et al. Targeting KRAS mutant cancers with a covalent G12C-specific inhibitor. Cell 172, 578–589.e17 (2018).

    Article  CAS  PubMed  Google Scholar 

  51. Kabsch, W. XDS. Acta Crystallogr. D Biol. Crystallogr. 66, 125–132 (2010).

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. 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  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  55. Auld, D. S. et al. "Assay Interference by Aggregation" in Assay Guidance Manual (eds Sittampalam, G. S. et al.) (Eli Lilly & Co. and the National Center for Advancing Translational Sciences, 2012).

  56. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

  57. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Xu, T. et al. ProLuCID: an improved SEQUEST-like algorithm with enhanced sensitivity and specificity. J. Proteomics 129, 16–24 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Sears, R., Ohtani, K. & Nevins, J. R. Identification of positively and negatively acting elements regulating expression of the E2F2 gene in response to cell growth signals. Mol. Cell. Biol. 17, 5227–5235 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Weiss, W. A., Aldape, K., Mohapatra, G., Feuerstein, B. G. & Bishop, J. M. Targeted expression of MYCN causes neuroblastoma in transgenic mice. EMBO J. 16, 2985–2995 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

N.L. is the incumbent of the Alan and Laraine Fischer Career Development Chair. N.L. thanks the Israel Science Foundation for funding (grant no. 2462/19), The Rising Tide Foundation, The Israel Cancer Research Fund, the Israeli Ministry of Science and Technology (grant no. 3-14763) and the Moross integrated cancer center. N.L. is also supported by the Helen and Martin Kimmel Center for Molecular Design, Joel and Mady Dukler Fund for Cancer Research, the Estate of Emile Mimran and Virgin JustGiving and the George Schwartzman Fund. C.D. was supported by the Minerva Fellowship program of the Max Planck Society, funded by the German Federal Ministry for Education and Research. This work was supported in part by NIH grant no. R01CA205153 to K.P.L., N.S.G. and X.Z.Z. N.S.G. was also supported by the Hale Center for Pancreatic Research. Y.C. and C.W. thank the Computing Platform of the Center for Life Science at Peking University for supporting proteomic data analysis. S.D.-P acknowledges funding from the Linde Family Foundation. Part of this research was conducted at the Advanced Photon Source on the Northeastern Collaborative Access Team beamlines (no. NIGMS P41 GM103403), and SBGrid compiled software (no. eLife 2013;2:e01456). J.A.M. acknowledges support from the National Institutes of Health (nos. CA233800 and TR002933) and the Mark Foundation for Cancer Research. B.J.P. was supported by the Ruth L. Kirschstein NRSA Individual Predoctoral Fellowship (no. F31 CA225066), the Training Grant in Pharmacological Sciences (no. NIH 5 T32 GM007306), the Training Grant in Chemical Biology (no. NIH 5 T32 GM095450-04) and the Chleck Foundation (also to Z.M.D.). B.N. was supported by an American Cancer Society Postdoctoral Fellowship (no. PF-17-010-01-CDD) and the Katherine L. and Steven C. Pinard Research Fund (also to N.S.G.). L.C. is supported by the Cancer Research UK Program Grant (nos. C34648/A18339 and C34648/A14610). Y.J. is supported by a Children with Cancer UK Research Fellowship (no. 2014/176). R.C.S. acknowledges funding support from NCI R01s (nos. CA196228 and CA186241) and foundation support from The Brenden-Colson Center for Pancreatic Care. We thank I. Ulitski for help with RNA-seq analysis, M. Kostic for critical reading of the manuscript, T.-M. Salame for help with FACS analysis and P. Gehrtz and I. You for helping with compound characterization.

Author information

Authors and Affiliations

Authors

Contributions

C.D., B.J.P., N.L. and N.S.G. wrote the manuscript with input from all authors. C.D. and T.D.M. undertook organic synthesis and design. C.D. performed covalent labeling studies. E.R. performed the covalent fragment screen. Chemoproteomics was carried out by Y.C., G.A., S. Sharifzadeh, S.B.F., C.M.B., C.W. and J.A.M. Crystallography was performed by H.-S.S., N.E.V., E.A.G. and S.D.-P. Pin1 biochemical assays were done by X.L., S. Kibe., S. Kozono. and B.J.P., led by X.Z.Z. and K.P.L. Pin1 cellular studies were performed by B.J.P., A.R., E.R., Z.M.D., E.K., T.D.M. and B.N. The PDAC mouse model is credited to K.K., led by X.Z.Z. and K.P.L. The Myc reporter is credited to E.M.L., C.J.D. and R.C.S. Neuroblastoma zebrafish models and cell lines are credited to S.H., M.W.Z. and T.L. Neuroblastoma mouse models are credited to E.P., Y.J., B.M.d.C. and L.C. Molecular modeling was performed by D.Z. The mouse PD study was conducted by A.Y. The mouse toxicity assay was performed by R.O. Radiation resensitization was carried out by R.B.S and S. Sidi. Germinal center studies were performed by L.S.-B. and Z.S. N.L. and N.S.G. conceived of and led this study.

Corresponding authors

Correspondence to Nathanael S. Gray or Nir London.

Ethics declarations

Competing interests

N.S.G. is a Scientific Founder and member of the Scientific Advisory Board (SAB) of C4, Jengu, Inception, Larkspur, Syros, Soltego, Gatekeeper and Petra Pharmaceuticals and has received research funding from Novartis, Astellas, Taiho and Deerfield. N.L. is a member of the SAB of Totus medicines and Monte Rosa Therapeutics and has received research support from Teva and Pfizer. J.A.M. has received support through sponsored research agreements with AstraZeneca and Vertex. J.A.M. serves on the SAB of 908 Devices. C.M.B. is an employee of AstraZeneca. C.D., B.J.P., D.Z., S.H., X.L., K.P.L., X.Z.Z., T.L., N.S.G. and N.L. are inventors on a patent application related to the inhibitors described in this manuscript (no. PCT/IL2020/050043).

Additional information

Peer review information Nature Chemical Biology thanks Timothy Chi-Wai Cheng, Yan Zhang and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Extended data

Extended Data Fig. 1 Chemoproteomic approaches to establish Sulfopin’s selectivity.

a, Schematic depiction of Covalent Inhibitor Target-Site Identification (CITe-Id) workflow, showing hypothetical results. CiTe-Id identifies Sulfopin-DTB modified sites across the proteome, and profiles competitively labeled cysteine residues following dose-response treatment with Sulfopin in live PATU-8988T cells. b, Schematic depicting the rdTOP-ABPP experimental workflow to assess Sulfopin proteomic selectivity in MDA-MB-231 cells.

Extended Data Fig. 2 Sulfopin phenocopies Pin1 knockout phenotypes.

a, HeLa cells were treated with either DMSO, Sulfopin, or Go6976 (a Chk1 inhibitor) and exposed to 7.5 Gy IR 1 h after drug treatment. Viability was assessed 3 days post-IR. Sulfopin shows a dose dependent sensitization of the cells to irradiation (n=3; data are represented as mean values with standard deviation). b, Western blot analysis was performed 24 h post-IR, showing Sulfopin blocked phosphorylation of Thr209 of IRAK1. c, A shorter exposure shows that Sulfopin inhibits IRAK1 phosphorylation already at concentrations of 0.1 μM. d, A scheme for testing the effect of Sulfopin in vivo on germinal center B cells in response to immunization. e, Representative flow cytometric plots with Vehicle and Sulfopin (left) and quantification (right) of FASHi CD38 germinal center (GC) cells in WT mice 11 days after immunization with NP-OVA. ** p<0.01, two tailed Student’s t test.

Source data

Extended Data Fig. 3 Sulfopin affects PATU-8988T cell cycle.

PATU-8988T cells were treated in triplicates or more for 4 days with either DMSO (0.1%), Sulfopin (2.5 M) or the non-covalent control Sulfopin-AcA (2.5 M). Cell cycle analysis was performed by BRDU and Propidium-Iodide staining, followed by FACS analysis. Sulfopin treatment reduces the % of cells in S-phase and in turn more cells are found in G1, while the non-covalent Sulfopin-AcA doesn’t show this effect. Representative FACS analysis graphs and a quantification of the results SD of two independent experiments are presented (A. n=3; B. n=4). Statistical significance was calculated using one-tailed Student’s t test (** = p < 0.01, *** = p < 0.001).

Extended Data Fig. 4 Sulfopin treatment does not induce apoptosis in cells.

a, PATU-8988T cells were treated for 5 or 6 days with either DMSO (0.1%), Sulfopin (1 μM, 2.5 μM) or the non-covalent control Sulfopin-AcA (2.5 μM). The cells were lysed and activation of caspase 3 and Pin1 levels were analysed by Western blot. As a positive control for caspase 3 activation the cells were treated with Staurosporin (1 μM, 4h; STS). See Supplementary Fig. 13a for the results of an additional independent experiment. Caspase 3 was not activated and Pin1 levels were not changed by the treatment with Sulfopin. b, PATU-8988T cells were treated in triplicates for 6 days with either DMSO (0.1%), Sulfopin (1 M, 2.5 M) or the non-covalent control Sulfopin-AcA (2.5 M). The cells were then stained with AnnexinV-FITC/ 7AAD and analysed by FACS. Staurosporin treatment (1 M, 4h) was used as a positive control for apoptosis. Representative FACS analysis graphs and a quantification of the results (n=3; data are represented as mean values with standard deviation). See Supplementary Fig. 13b for the results of an additional independent experiment. Live cells were defined as AnnexinV-/7AAD-, early apoptosis AnnexinV+/7AAD- and late apoptosis AnnexinV+/ 7AAD+.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–15, Tables 1–4 and Note (synthetic procedures and analytical data).

Reporting Summary

Supplementary Datasets

Supplementary Datasets 1–6.

Source data

Source Data Fig. 2

Unprocessed immunolots.

Source Data Extended Data Fig. 2

Unprocessed immunolots.

Source Data Extended Data Fig. 4

Unprocessed immunolots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dubiella, C., Pinch, B.J., Koikawa, K. et al. Sulfopin is a covalent inhibitor of Pin1 that blocks Myc-driven tumors in vivo. Nat Chem Biol 17, 954–963 (2021). https://doi.org/10.1038/s41589-021-00786-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-021-00786-7

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research