Abstract

Abstract Metastatic castration resistance prostate cancer (mCRPC) is a lethal disease due to the development of treatment to androgen receptor (AR) signaling inhibitors (ARSi) and taxane chemotherapy. Treatment resistance occurs partly due to the expression of AR splice variants that lack the ligand binding domain (LBD) and are constitutively active in the nucleus. AR-V7 is the most prevalent variant conferring clinical resistance to both ARSi and taxanes. Currently, there is no selective AR-V7 inhibitor leaving patients with limited therapeutic options. Thus, the development of selective AR-V7 inhibitors is a high priority, clinically unmet need. To identify AR-V7 pharmacologic inhibitors, we performed a high throughput small molecule phenotypic screen using enzymatic complementation and nuclear AR-V7 as the assay endpoint. Our primary screen of ~170K compounds (z score = 0.8), followed by a cell-toxicity counter screen and a secondary GFP screen identified hit compound 7907, as a dual AR-V7/AR-fl protein degrader, with unique chemotype compared to all known AR modulators. Hit to lead optimization by medicinal chemistry/SAR studies identified lead compound 15, with increased potency compared to 7907. Mechanistically, we showed that compound 15 shortened AR-V7 protein half-life by activating the ubiquitin-proteasome pathway and inducing proteasomal degradation of both AR-V7/AR-fl, without affecting their transcription. Importantly, compound 15 induced degradation occurred within 3hr of treatment and was blocked by the clinically approved proteasome inhibitor, bortezomib. TurboID proximity ligation assay identified distinct E3 ligases, uniquely interacting with AR-V7 or AR-fl. Using AR-V7/AR-fl deletion mutants we further showed that compound 15 activity is mediated by the N-terminal domain of AR, present in both proteins. Remarkably, compound 15 sensitized LNCaP95 cells (endogenous AR-V7/AR-fl expression) to enzalutamide suggesting potential therapeutic synergism and ability to reverse enzalutamide resistance. Ongoing studies aim to narrow down the binding site of compound 15 as well as identify the E3 ligases mediating its activity. Together, these data support a molecular glue degrader mechanism of action, consistent with published studies showing that molecular glue degraders are ideal for targeting classically “undruggable” proteins lacking an LBD or containing intrinsically disordered domains, as is the case for AR-V7. Currently, all AR-directed therapies target the LBD of AR-fl, inhibiting AR signaling. AR-V7 expression is a direct outcome of this inhibition, leading to AR-fl and AR-V7 co-expression in patient tumors. We posit that our drug candidate, by offering dual AR-V7/AR-fl inhibition in a single treatment, has the potential to not only benefit patients with mCRPC but also patients with hormone-sensitive disease, and delay AR-V7 expression. Citation Format: CheukMan C. Au, Catrina Estrella, Prerna Vatsa, Michelle Naidoo, Michael Miller, Peter T. Meinke, David M. Nanus, Paraskevi Giannakakou. First-in-class AR-V7/AR-fl small molecule molecular glue degrader for prostate cancer treatment [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2024; Part 1 (Regular Abstracts); 2024 Apr 5-10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2024;84(6_Suppl):Abstract nr 1231.

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