The prolyl isomerase PIN1, a critical modifier of multiple signalling pathways, is overexpressed in the majority of cancers and its activity strongly contributes to tumour initiation and progression. Inactivation of PIN1 function conversely curbs tumour growth and cancer stem cell expansion, restores chemosensitivity and blocks metastatic spread, thus providing the rationale for a therapeutic strategy based on PIN1 inhibition. Notwithstanding, potent PIN1 inhibitors are still missing from the arsenal of anti-cancer drugs. By a mechanism-based screening, we have identified a novel covalent PIN1 inhibitor, KPT-6566, able to selectively inhibit PIN1 and target it for degradation. We demonstrate that KPT-6566 covalently binds to the catalytic site of PIN1. This interaction results in the release of a quinone-mimicking drug that generates reactive oxygen species and DNA damage, inducing cell death specifically in cancer cells. Accordingly, KPT-6566 treatment impairs PIN1-dependent cancer phenotypes in vitro and growth of lung metastasis in vivo.

A covalent PIN1 inhibitor selectively targets cancer cells by a dual mechanism of action

Campaner, Elena;Rustighi, Alessandra;Zannini, Alessandro;Cristiani, Alberto;Piazza, Silvano;Ciani, Yari;Giabbai, Barbara;Storici, Paola;D'Incalci, Maurizio;Rosato, Antonio;Del Sal, Giannino
2017-01-01

Abstract

The prolyl isomerase PIN1, a critical modifier of multiple signalling pathways, is overexpressed in the majority of cancers and its activity strongly contributes to tumour initiation and progression. Inactivation of PIN1 function conversely curbs tumour growth and cancer stem cell expansion, restores chemosensitivity and blocks metastatic spread, thus providing the rationale for a therapeutic strategy based on PIN1 inhibition. Notwithstanding, potent PIN1 inhibitors are still missing from the arsenal of anti-cancer drugs. By a mechanism-based screening, we have identified a novel covalent PIN1 inhibitor, KPT-6566, able to selectively inhibit PIN1 and target it for degradation. We demonstrate that KPT-6566 covalently binds to the catalytic site of PIN1. This interaction results in the release of a quinone-mimicking drug that generates reactive oxygen species and DNA damage, inducing cell death specifically in cancer cells. Accordingly, KPT-6566 treatment impairs PIN1-dependent cancer phenotypes in vitro and growth of lung metastasis in vivo.
2017
Pubblicato
https://www.nature.com/articles/ncomms15772
File in questo prodotto:
File Dimensione Formato  
campaner2017.pdf

accesso aperto

Tipologia: Documento in Versione Editoriale
Licenza: Creative commons
Dimensione 1.31 MB
Formato Adobe PDF
1.31 MB Adobe PDF Visualizza/Apri
ncomms15772-s1.pdf

accesso aperto

Descrizione: supplementary informations
Tipologia: Altro materiale allegato
Licenza: Creative commons
Dimensione 2.85 MB
Formato Adobe PDF
2.85 MB Adobe PDF Visualizza/Apri
ncomms15772-s2.pdf

accesso aperto

Descrizione: supplementary informations
Tipologia: Altro materiale allegato
Licenza: Creative commons
Dimensione 343.28 kB
Formato Adobe PDF
343.28 kB Adobe PDF Visualizza/Apri
Pubblicazioni consigliate

I documenti in IRIS sono protetti da copyright e tutti i diritti sono riservati, salvo diversa indicazione.

Utilizza questo identificativo per citare o creare un link a questo documento: https://hdl.handle.net/11368/2919875
Citazioni
  • ???jsp.display-item.citation.pmc??? 48
  • Scopus 96
  • ???jsp.display-item.citation.isi??? 92
social impact