Your browser doesn't support javascript.
loading
Drug-Induced Epigenomic Plasticity Reprograms Circadian Rhythm Regulation to Drive Prostate Cancer toward Androgen Independence.
Linder, Simon; Hoogstraat, Marlous; Stelloo, Suzan; Eickhoff, Nils; Schuurman, Karianne; de Barros, Hilda; Alkemade, Maartje; Bekers, Elise M; Severson, Tesa M; Sanders, Joyce; Huang, Chia-Chi Flora; Morova, Tunc; Altintas, Umut Berkay; Hoekman, Liesbeth; Kim, Yongsoo; Baca, Sylvan C; Sjöström, Martin; Zaalberg, Anniek; Hintzen, Dorine C; de Jong, Jeroen; Kluin, Roelof J C; de Rink, Iris; Giambartolomei, Claudia; Seo, Ji-Heui; Pasaniuc, Bogdan; Altelaar, Maarten; Medema, René H; Feng, Felix Y; Zoubeidi, Amina; Freedman, Matthew L; Wessels, Lodewyk F A; Butler, Lisa M; Lack, Nathan A; van der Poel, Henk; Bergman, Andries M; Zwart, Wilbert.
Afiliação
  • Linder S; Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
  • Hoogstraat M; Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
  • Stelloo S; Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
  • Eickhoff N; Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
  • Schuurman K; Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
  • de Barros H; Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
  • Alkemade M; Division of Urology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
  • Bekers EM; Core Facility Molecular Pathology and Biobanking, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
  • Severson TM; Division of Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
  • Sanders J; Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
  • Huang CF; Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
  • Morova T; Division of Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
  • Altintas UB; Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada.
  • Hoekman L; Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada.
  • Kim Y; School of Medicine, Koç University, Istanbul, Turkey.
  • Baca SC; Koç University Research Centre for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey.
  • Sjöström M; Proteomics Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
  • Zaalberg A; Department of Pathology, Amsterdam University Medical Centers, Cancer Center Amsterdam, Amsterdam, the Netherlands.
  • Hintzen DC; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
  • de Jong J; The Eli and Edythe L. Broad Institute, Cambridge, Massachusetts.
  • Kluin RJC; Department of Radiation Oncology, University of California, San Francisco, San Francisco, California.
  • de Rink I; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California.
  • Giambartolomei C; Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
  • Seo JH; Division of Cell Biology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
  • Pasaniuc B; Division of Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
  • Altelaar M; Genomics Core Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
  • Medema RH; Genomics Core Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
  • Feng FY; Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.
  • Zoubeidi A; Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.
  • Freedman ML; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
  • Wessels LFA; Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.
  • Butler LM; Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.
  • Lack NA; Proteomics Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
  • van der Poel H; Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht Institute for Pharmaceutical Sciences, Utrecht University and Netherlands Proteomics Centre, Utrecht, the Netherlands.
  • Bergman AM; Division of Cell Biology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
  • Zwart W; Department of Radiation Oncology, University of California, San Francisco, San Francisco, California.
Cancer Discov ; 12(9): 2074-2097, 2022 09 02.
Article em En | MEDLINE | ID: mdl-35754340
ABSTRACT
In prostate cancer, androgen receptor (AR)-targeting agents are very effective in various disease stages. However, therapy resistance inevitably occurs, and little is known about how tumor cells adapt to bypass AR suppression. Here, we performed integrative multiomics analyses on tissues isolated before and after 3 months of AR-targeting enzalutamide monotherapy from patients with high-risk prostate cancer enrolled in a neoadjuvant clinical trial. Transcriptomic analyses demonstrated that AR inhibition drove tumors toward a neuroendocrine-like disease state. Additionally, epigenomic profiling revealed massive enzalutamide-induced reprogramming of pioneer factor FOXA1 from inactive chromatin sites toward active cis-regulatory elements that dictate prosurvival signals. Notably, treatment-induced FOXA1 sites were enriched for the circadian clock component ARNTL. Posttreatment ARNTL levels were associated with patients' clinical outcomes, and ARNTL knockout strongly decreased prostate cancer cell growth. Our data highlight a remarkable cistromic plasticity of FOXA1 following AR-targeted therapy and revealed an acquired dependency on the circadian regulator ARNTL, a novel candidate therapeutic target.

SIGNIFICANCE:

Understanding how prostate cancers adapt to AR-targeted interventions is critical for identifying novel drug targets to improve the clinical management of treatment-resistant disease. Our study revealed an enzalutamide-induced epigenomic plasticity toward prosurvival signaling and uncovered the circadian regulator ARNTL as an acquired vulnerability after AR inhibition, presenting a novel lead for therapeutic development. See related commentary by Zhang et al., p. 2017. This article is highlighted in the In This Issue feature, p. 2007.
Assuntos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Neoplasias de Próstata Resistentes à Castração / Androgênios Tipo de estudo: Prognostic_studies Limite: Humans / Male Idioma: En Ano de publicação: 2022 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Neoplasias de Próstata Resistentes à Castração / Androgênios Tipo de estudo: Prognostic_studies Limite: Humans / Male Idioma: En Ano de publicação: 2022 Tipo de documento: Article