The androgen receptor (AR) signaling pathway is the key driver of human prostate cancer. Prostate cancers are almost always sensitive to blockade of the androgen receptor, and tumors with acquired resistance to these therapies are often sensitive to second-generation inhibitors of the pathway. However, the vast majority of men with progressive prostate cancer will ultimately succumb to the disease. Activation of the PI3K signaling pathway is the second most prevalent oncogenic event in prostate cancer, occurring in the majority of advanced castration-resistant tumors. The PTEN tumor suppressor gene is lost or inactivated in 40% of these tumors, and genetic events that activate PI3K are also found in a significant number of patients. While PTEN loss is the most commonly altered regulator of PI3K signaling in prostate cancer, recent genomic profiling studies of metastatic castration-resistant prostate cancer (CRPC) have reported activating alterations in the subunits of the PI3K complex (PIK3CA, PIK3CB, PIK3R1) in approximately 10% to 20% of cases. Our previous work revealed that activation of AR-signaling or PI3K signaling each cause feedback inhibition of the other in a reciprocal fashion. As predicted from this finding, inhibition of androgen receptor signaling activates PI3K signaling in these tumors and correspondingly, inhibitors of PI3K signaling activate AR signaling. It is likely that relief of feedback inhibition of the other pathway reduces the benefit of therapy with single agent AR or PI3K inhibitors. In support of this hypothesis, we showed that combined inhibition of both pathways has profound therapeutic effects in preclinical models of advanced prostate cancer. The major goal of this proposal is to optimize such therapies, tailoring the therapeutic strategy to specific tumor genotypes. We will do this by determining the biologic consequences of different mechanisms of PI3K activation in prostate cancer, determining the best means of pharmacologically inhibiting PI3K and AR signaling in prostate tumors with PTEN loss and/or PI3K activation, and optimizing the dose and schedule of these combinations in novel organoid and genetically engineered mouse models of prostate cancer.
The aims of this proposal are to explore the biologic consequences of PI3K pathway activation in AR-driven prostate cancer and to determine the optimal combination regimens with which to treat these tumors.

Public Health Relevance

Prostate cancers are almost always sensitive to blockade of the androgen receptor (AR), and tumors with acquired resistance to these therapies are often sensitive to second-generation inhibitors of the pathway. However, the vast majority of men with progressive prostate cancer will ultimately succumb to the disease. We propose to optimize the use of PI3K pathway inhibitors in prostate cancer by determining how the PI3K mutational landscape impacts dependency towards p110 isoform-selective inhibitors. The knowledge gained from this project will provide us with the greatest chance for rapidly developing a rationally designed clinical trial evaluating the combined inhibition of the PI3K and AR pathways in men with prostate cancer to maximize therapeutic efficacy and minimize associated toxicity.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Specialized Center (P50)
Project #
5P50CA092629-19
Application #
9774813
Study Section
Special Emphasis Panel (ZCA1)
Project Start
Project End
Budget Start
2019-09-01
Budget End
2020-08-31
Support Year
19
Fiscal Year
2019
Total Cost
Indirect Cost
Name
Sloan-Kettering Institute for Cancer Research
Department
Type
DUNS #
064931884
City
New York
State
NY
Country
United States
Zip Code
10065
Han, SoHyun; Stoyanova, Radka; Lee, Hansol et al. (2018) Automation of pattern recognition analysis of dynamic contrast-enhanced MRI data to characterize intratumoral vascular heterogeneity. Magn Reson Med 79:1736-1744
Vickers, Andrew J; Steineck, Gunnar (2018) Prognosis, Effect Modification, and Mediation. Eur Urol 74:243-245
Kinsella, Netty; Helleman, Jozien; Bruinsma, Sophie et al. (2018) Active surveillance for prostate cancer: a systematic review of contemporary worldwide practices. Transl Androl Urol 7:83-97
Hieronymus, Haley; Murali, Rajmohan; Tin, Amy et al. (2018) Tumor copy number alteration burden is a pan-cancer prognostic factor associated with recurrence and death. Elife 7:
Scher, Howard I; Graf, Ryon P; Schreiber, Nicole A et al. (2018) Assessment of the Validity of Nuclear-Localized Androgen Receptor Splice Variant 7 in Circulating Tumor Cells as a Predictive Biomarker for Castration-Resistant Prostate Cancer. JAMA Oncol 4:1179-1186
Bielski, Craig M; Zehir, Ahmet; Penson, Alexander V et al. (2018) Genome doubling shapes the evolution and prognosis of advanced cancers. Nat Genet 50:1189-1195
Luo, Jun; Attard, Gerhardt; Balk, Steven P et al. (2018) Role of Androgen Receptor Variants in Prostate Cancer: Report from the 2017 Mission Androgen Receptor Variants Meeting. Eur Urol 73:715-723
Settleman, Jeffrey; Sawyers, Charles L; Hunter, Tony (2018) Challenges in validating candidate therapeutic targets in cancer. Elife 7:
Miyazawa, Miki; Subbaramaiah, Kotha; Bhardwaj, Priya et al. (2018) Pioglitazone Inhibits Periprostatic White Adipose Tissue Inflammation in Obese Mice. Cancer Prev Res (Phila) 11:215-226
Graham, Laura; Banda, Kalyan; Torres, Alba et al. (2018) A phase II study of the dual mTOR inhibitor MLN0128 in patients with metastatic castration resistant prostate cancer. Invest New Drugs 36:458-467

Showing the most recent 10 out of 505 publications