4.7 Article

BET Bromodomain Inhibition Blocks an AR-Repressed, E2F1-Activated Treatment-Emergent Neuroendocrine Prostate Cancer Lineage Plasticity Program

Journal

CLINICAL CANCER RESEARCH
Volume 27, Issue 17, Pages 4923-4936

Publisher

AMER ASSOC CANCER RESEARCH
DOI: 10.1158/1078-0432.CCR-20-4968

Keywords

-

Categories

Funding

  1. NCI [R01 CA251245, R01CA234715]
  2. NCI Pacific Northwest Prostate Cancer SPORE [P50 CA097186]
  3. NCI Michigan Prostate SPORE [P50 CA186786]
  4. NCI Drug Resistance and Sensitivity Network [P50 CA186786-07S1, U54CA224079]
  5. Department of Defense Impact Award [W81XWH-16-1-0597]
  6. Department of Defense Prostate Cancer Research Program Physician Research Award [W81XWH-17-1-0124]
  7. Prostate Cancer Foundation Young Investigator Award
  8. Kuni Foundation
  9. University of Michigan Rogel Scholar Award
  10. Rogel Innovation Award/NCI [P30 CA046592]
  11. Sheppard Family Fund Sheppard Scholar Award
  12. Zenith Epigenetics

Ask authors/readers for more resources

NEPC is the most aggressive lineage plasticity in prostate cancer, with limited treatment options. By inhibiting AR and using BET inhibitors, it is possible to block this lineage plasticity, showing promising potential for t-NEPC treatment.
Purpose: Lineage plasticity in prostate cancer-most commonly exemplified by loss of androgen receptor (AR) signaling and a switch from a luminal to alternate differentiation program-is now recognized as a treatment resistance mechanism. Lineage plasticity is a spectrum, but neuroendocrine prostate cancer (NEPC) is the most virulent example. Currently, there are limited treatments for NEPC. Moreover, the incidence of treatment-emergent NEPC (t-NEPC) is increasing in the era of novel AR inhibitors. In contradistinction to de novo NEPC, t-NEPC tumors often express the AR, but AR's functional role in t-NEPC is unknown. Furthermore, targetable factors that promote t-NEPC lineage plasticity are also unclear. Experimental Design: Using an integrative systems biology approach, we investigated enzalutamide-resistant t-NEPC cell lines and their parental, enzalutamide-sensitive adenocarcinoma cell lines. The AR is still expressed in these t-NEPC cells, enabling us to determine the role of the AR and other key factors in regulating t-NEPC lineage plasticity. Results: AR inhibition accentuates lineage plasticity in t-NEPC cells-an effect not observed in parental, enzalutamide-sensitive adenocarcinoma cells. Induction of an AR-repressed, lineage plasticity program is dependent on activation of the transcription factor E2F1 in concert with the BET bromodomain chromatin reader BRD4. BET inhibition (BETi) blocks this E2F1/BRD4-regulated program and decreases growth of t-NEPC tumor models and a subset of t-NEPC patient tumors with high activity of this program in a BETi clinical trial. Conclusions: E2F1 and BRD4 are critical for activating an AR-repressed, t-NEPC lineage plasticity program. BETi is a promising approach to block this program.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available