Oncotarget

Research Papers:

PACE4 inhibitors and their peptidomimetic analogs block prostate cancer tumor progression through quiescence induction, increased apoptosis and impaired neovascularisation

Christine Levesque _, Frédéric Couture, Anna Kwiatkowska, Roxane Desjardins, Brigitte Guérin, Witold A. Neugebauer and Robert Day

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2015; 6:3680-3693. https://doi.org/10.18632/oncotarget.2918

Metrics: PDF 2312 views  |   HTML 3744 views  |   ?  


Abstract

Christine Levesque1,2, Frédéric Couture1,2, Anna Kwiatkowska1,2, Roxane Desjardins1,2, Brigitte Guérin2,3, Witold A. Neugebauer2, Robert Day1,2

1Department of Surgery/Urology Division and Faculté de Médecine et Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec, J1H 5N4, Canada

2Institut de Pharmacologie de Sherbrooke, Faculté de Médecine et Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec, J1H 5N4, Canada

3Department of Nuclear Medicine and Radiobiology and Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Faculté de Médecine et Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec, J1H 5N4, Canada

Correspondence to:

Robert Day, e-mail: [email protected]

Keywords: PACE4, Multi-Leu peptide, Prostate Cancer, Proprotein convertases, Peptide inhibitors

Received: November 16, 2014     Accepted: December 14, 2014     Published: February 19, 2015

ABSTRACT

Prostate cancer is the leading cancer in North American men. Current pharmacological treatments are limited to anti-androgen strategies and the development of new therapeutic approaches remains a challenge. As a fundamentally new approach, we propose the inhibition of PACE4, a member of the proprotein convertases family of enzymes, as a therapeutic target in prostate cancer. We developed an inhibitor named the Multi-Leu peptide, with potent in vitro anti-proliferative effects. However, the Multi-Leu peptide has not been tested under in vivo conditions and its potency under such conditions is most likely limited, due to the labile characteristics of peptides in general. Using a peptidomimetic approach, we modified the initial scaffold, generating the analog Ac-[DLeu]LLLRVK-Amba, which demonstrates increased inhibitory potency and stability. The systemic administration of this peptidomimetic significantly inhibits tumor progression in the LNCaP xenograft model of prostate cancer by inducing tumor cell quiescence, increased apoptosis and neovascularization impairment. Pharmacokinetic and biodistribution profiles of this inhibitor confirm adequate tumor delivery properties of the compound. We conclude that PACE4 peptidomimetic inhibitors could result in stable and potent drugs for a novel therapeutic strategy for prostate cancer.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 2918