Abstract

Abstract Prostate tumors initially respond to androgen deprivation therapy but eventually progress despite low serum levels of testosterone. In this stage, known as castration-resistant prostate cancer (CRPC), abnormal androgen receptor (AR) signaling persists and classic anti-androgens such as bicalutamide are ineffective at controlling tumor growth. The novel AR signaling inhibitor MDV3100 has been demonstrated to inhibit AR signaling in preclinical models of CRPC. Here, we investigate further the cellular effects of MDV3100 treatment on AR signaling in in vitro models and in vivo xenograft models of CRPC. Our experiments reveal that MDV3100 (> 100 nM) inhibits dihydrotestosterone (DHT) induced AR-target gene expression in LNCaP cells and LNCaP/AR cells, a CRPC cell culture model in which AR is over-expressed. Inhibition of Prostate-Specific Antigen mRNA expression by MDV3100 was effective at concentrations 10-fold lower than with bicalutamide. As evidenced in other studies and in contrast to bicalutamide, MDV3100 did not display agonistic effects on AR signaling at 10 μM. In contrast to bicalutamide, MDV3100 (> 1 μM) inhibited androgen dependent nuclear translocation of AR, as measured with a quantitative optical based enzyme fragment complementation assay and AR-YFP localization in HEK-293 cells. Furthermore, in the presence of DHT, MDV3100 effectively reduced cell viability of LNCaP cells at concentrations as low as 0.1 μM, 10-fold lower than bicalutamide. MDV3100 also decreased cell viability of LNCaP/AR and W741C-LNcaP cells. We found that the apoptosis marker cleaved-caspase 3 was induced in LNCaP/AR cells treated with 1 M MDV3100 for 48 hours. Under these conditions, MDV3100 was more potent than bicalutamide in decreasing cell viability. Finally, we show that MDV3100 inhibited tumor growth and induced regression of tumor volume in a xenograft model of CRPC generated with LNCaP/AR cells. These results demonstrate that MDV3100, unlike anti-androgens, is effective in both in vitro and in vivo models of CRPC and that its mechanistic advantages over other anti-androgens could be explained by its ability to inhibit AR nuclear translocation and induce cell death. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2011 Nov 12-16; San Francisco, CA. Philadelphia (PA): AACR; Mol Cancer Ther 2011;10(11 Suppl):Abstract nr A76.

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call