Your browser doesn't support javascript.
loading
BET Bromodomain Inhibition Blocks an AR-Repressed, E2F1-Activated Treatment-Emergent Neuroendocrine Prostate Cancer Lineage Plasticity Program.
Kim, Dae-Hwan; Sun, Duanchen; Storck, William K; Welker Leng, Katherine; Jenkins, Chelsea; Coleman, Daniel J; Sampson, David; Guan, Xiangnan; Kumaraswamy, Anbarasu; Rodansky, Eva S; Urrutia, Joshua A; Schwartzman, Jacob A; Zhang, Chao; Beltran, Himisha; Labrecque, Mark P; Morrissey, Colm; Lucas, Jared M; Coleman, Ilsa M; Nelson, Peter S; Corey, Eva; Handelman, Samuel K; Sexton, Jonathan Z; Aggarwal, Rahul; Abida, Wassim; Feng, Felix Y; Small, Eric J; Spratt, Daniel E; Bankhead, Armand; Rao, Arvind; Gesner, Emily M; Attwell, Sarah; Lakhotia, Sanjay; Campeau, Eric; Yates, Joel A; Xia, Zheng; Alumkal, Joshi J.
Afiliação
  • Kim DH; Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, Oregon.
  • Sun D; Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, Oregon.
  • Storck WK; Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan.
  • Welker Leng K; Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
  • Jenkins C; Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan.
  • Coleman DJ; Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
  • Sampson D; Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, Oregon.
  • Guan X; Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, Oregon.
  • Kumaraswamy A; Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, Oregon.
  • Rodansky ES; Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, Oregon.
  • Urrutia JA; Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan.
  • Schwartzman JA; Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
  • Zhang C; Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan.
  • Beltran H; Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
  • Labrecque MP; Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, Oregon.
  • Morrissey C; Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, Oregon.
  • Lucas JM; Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan.
  • Coleman IM; Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
  • Nelson PS; Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
  • Corey E; Department of Urology, University of Washington, Seattle, Washington.
  • Handelman SK; Department of Urology, University of Washington, Seattle, Washington.
  • Sexton JZ; Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington.
  • Aggarwal R; Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington.
  • Abida W; Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington.
  • Feng FY; Department of Urology, University of Washington, Seattle, Washington.
  • Small EJ; Center for Drug Repurposing, Department of Internal Medicine, Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan.
  • Spratt DE; Center for Drug Repurposing, Department of Internal Medicine, Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan.
  • Bankhead A; Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California.
  • Rao A; Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
  • Gesner EM; Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California.
  • Attwell S; Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California.
  • Lakhotia S; Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
  • Campeau E; Department of Radiation Oncology, University Hospitals, Case Western Reserve University, Cleveland, Ohio.
  • Yates JA; Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
  • Xia Z; Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, Michigan.
  • Alumkal JJ; Department of Biostatistics, University of Michigan, Ann Arbor, Michigan.
Clin Cancer Res ; 27(17): 4923-4936, 2021 09 01.
Article em En | MEDLINE | ID: mdl-34145028
ABSTRACT

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.
Assuntos

Texto completo: 1 Coleções: 01-internacional Temas: Geral / Tipos_de_cancer / Prostata Base de dados: MEDLINE Assunto principal: Feniltioidantoína / Neoplasias da Próstata / Benzamidas / Proteínas / Carcinoma Neuroendócrino / Fator de Transcrição E2F1 / Antagonistas de Receptores de Andrógenos / Antineoplásicos / Nitrilas Tipo de estudo: Prognostic_studies Limite: Humans / Male Idioma: En Revista: Clin Cancer Res Assunto da revista: NEOPLASIAS Ano de publicação: 2021 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Temas: Geral / Tipos_de_cancer / Prostata Base de dados: MEDLINE Assunto principal: Feniltioidantoína / Neoplasias da Próstata / Benzamidas / Proteínas / Carcinoma Neuroendócrino / Fator de Transcrição E2F1 / Antagonistas de Receptores de Andrógenos / Antineoplásicos / Nitrilas Tipo de estudo: Prognostic_studies Limite: Humans / Male Idioma: En Revista: Clin Cancer Res Assunto da revista: NEOPLASIAS Ano de publicação: 2021 Tipo de documento: Article