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

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 ac...

Full description

Saved in:
Bibliographic Details
Published inNature chemical biology Vol. 17; no. 9; pp. 954 - 963
Main Authors Dubiella, Christian, Pinch, Benika J., Koikawa, Kazuhiro, Zaidman, Daniel, Poon, Evon, Manz, Theresa D., Nabet, Behnam, He, Shuning, Resnick, Efrat, Rogel, Adi, Langer, Ellen M., Daniel, Colin J., Seo, Hyuk-Soo, Chen, Ying, Adelmant, Guillaume, Sharifzadeh, Shabnam, Ficarro, Scott B., Jamin, Yann, Martins da Costa, Barbara, Zimmerman, Mark W., Lian, Xiaolan, Kibe, Shin, Kozono, Shingo, Doctor, Zainab M., Browne, Christopher M., Yang, Annan, Stoler-Barak, Liat, Shah, Richa B., Vangos, Nicholas E., Geffken, Ezekiel A., Oren, Roni, Koide, Eriko, Sidi, Samuel, Shulman, Ziv, Wang, Chu, Marto, Jarrod A., Dhe-Paganon, Sirano, Look, Thomas, Zhou, Xiao Zhen, Lu, Kun Ping, Sears, Rosalie C., Chesler, Louis, Gray, Nathanael S., London, Nir
Format Journal Article
LanguageEnglish
Published New York Nature Publishing Group US 01.09.2021
Nature Publishing Group
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary: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. The development of Sulfopin, a highly selective and potent, covalent Pin1 inhibitor that phenocopies Pin1 knockout and regresses tumors in murine and zebrafish models of neuroblastoma as well as in a pancreatic cancer mouse model.
Bibliography: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.
Author contributions
ISSN:1552-4450
1552-4469
DOI:10.1038/s41589-021-00786-7