A Druggable FOXA1-Glucocorticoid Receptor Transcriptional Axis Drives Tumor Growth in a Subset of Non-Small Cell Lung Cancer

Author:

Dorso M.12ORCID,Patel Payal T.12ORCID,Pankov Aleksandr3ORCID,Boyer Jacob A.4ORCID,Soni Rajesh K.5ORCID,Del Priore Isabella S.1ORCID,Hayatt Omar6ORCID,Kulick Amanda6ORCID,Hagen Connor J.6ORCID,de Stanchina Elisa6ORCID,Junttila Melissa R.3ORCID,Daemen Anneleen3ORCID,Friedman Lori S.3ORCID,Hendrickson Ronald C.5ORCID,Chandarlapaty Sarat17ORCID

Affiliation:

1. 1Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.

2. 2Pharmacology Graduate Program, Weill Cornell Medicine, New York, New York.

3. 3ORIC Pharmaceuticals, South San Francisco, California.

4. 4Gerstner Sloan Kettering Graduate Program, Sloan Kettering Institute, New York, New York.

5. 5Microchemistry and Proteomics Core, Sloan Kettering Institute, New York, New York.

6. 6Antitumor Assessment Core, Memorial Sloan Kettering Cancer Center, New York, New York.

7. 7Breast Medicine Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.

Abstract

The FOXA1 pioneer factor is an essential mediator of steroid receptor function in multiple hormone-dependent cancers, including breast and prostate cancers, enabling nuclear receptors such as estrogen receptor (ER) and androgen receptor (AR) to activate lineage-specific growth programs. FOXA1 is also highly expressed in non–small cell lung cancer (NSCLC), but whether and how it regulates tumor growth in this context is not known. Analyzing data from loss-of-function screens, we identified a subset of NSCLC tumor lines where proliferation is FOXA1 dependent. Using rapid immunoprecipitation and mass spectrometry of endogenous protein, we identified chromatin-localized interactions between FOXA1 and glucocorticoid receptor (GR) in these tumor cells. Knockdown of GR inhibited proliferation of FOXA1-dependent, but not FOXA1-independent NSCLC cells. In these FOXA1-dependent models, FOXA1 and GR cooperate to regulate gene targets involved in EGF signaling and G1–S cell-cycle progression. To investigate the therapeutic potential for targeting this complex, we examined the effects of highly selective inhibitors of the GR ligand-binding pocket and found that GR antagonism with ORIC-101 suppressed FOXA1/GR target expression, activation of EGF signaling, entry into the S-phase, and attendant proliferation in vitro and in vivo. Taken together, our findings point to a subset of NSCLCs harboring a dependence on the FOXA1/GR growth program and provide rationale for its therapeutic targeting. Significance: NSCLC is the leading cause of cancer deaths worldwide. There is a need to identify novel druggable dependencies. We identify a subset of NSCLCs dependent on FOXA1-GR and sensitive to GR antagonism.

Funder

HHS | National Institutes of Health

Breast Cancer Research Foundation

Cancer Couch Foundation

Publisher

American Association for Cancer Research (AACR)

Reference57 articles.

1. Transcription factors and chromatin proteins as therapeutic targets in cancer;Johnston;Biochim Biophys Acta,2015

2. Biological subtypes of breast cancer: Prognostic and therapeutic implications;Yersal;World J Clin Oncol,2014

3. Androgen receptor in prostate cancer;Heinlein;Endocr Rev,2004

4. Tamoxifen: a most unlikely pioneering medicine;Jordan;Nat Rev Drug Discov,2003

5. Fulvestrant – a novel endocrine therapy for breast cancer;Johnston;Curr Med Chem,2010

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3