Oncotarget

Priority Research Papers:

Progesterone receptor isoforms, agonists and antagonists differentially reprogram estrogen signaling

Hari Singhal, Marianne E. Greene, Allison L. Zarnke, Muriel Laine, Rose Al Abosy, Ya-Fang Chang, Anna G. Dembo, Kelly Schoenfelt, Raga Vadhi, Xintao Qiu, Prakash Rao, Bindu Santhamma, Hareesh B. Nair, Klaus J. Nickisch, Henry W. Long, Lev Becker, Myles Brown and Geoffrey L. Greene _

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2018; 9:4282-4300. https://doi.org/10.18632/oncotarget.21378

Metrics: PDF 2593 views  |   HTML 6146 views  |   ?  


Abstract

Hari Singhal1, Marianne E. Greene3, Allison L. Zarnke3, Muriel Laine3, Rose Al Abosy3, Ya-Fang Chang3, Anna G. Dembo3, Kelly Schoenfelt3, Raga Vadhi2, Xintao Qiu2, Prakash Rao2, Bindu Santhamma4, Hareesh B. Nair4, Klaus J. Nickisch4, Henry W. Long2, Lev Becker3, Myles Brown1,2 and Geoffrey L. Greene3

1 Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA

2 Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts, USA

3 Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois, USA

4 Evestra Inc., San Antonio, Texas, USA

Correspondence to:

Geoffrey L. Greene, email:

Keywords: cancer, hormones, estrogen, progesterone

Received: August 03, 2017 Accepted: September 05, 2017 Published: September 28, 2017

Abstract

Major roadblocks to developing effective progesterone receptor (PR)-targeted therapies in breast cancer include the lack of highly-specific PR modulators, a poor understanding of the pro- or anti-tumorigenic networks for PR isoforms and ligands, and an incomplete understanding of the cross talk between PR and estrogen receptor (ER) signaling. Through genomic analyses of xenografts treated with various clinically-relevant ER and PR-targeting drugs, we describe how the activation or inhibition of PR differentially reprograms estrogen signaling, resulting in the segregation of transcriptomes into separate PR agonist and antagonist-mediated groups. These findings address an ongoing controversy regarding the clinical utility of PR agonists and antagonists, alone or in combination with tamoxifen, for breast cancer management. Additionally, the two PR isoforms PRA and PRB, bind distinct but overlapping genomic sites and interact with different sets of co-regulators to differentially modulate estrogen signaling to be either pro- or anti-tumorigenic. Of the two isoforms, PRA inhibited gene expression and ER chromatin binding significantly more than PRB. Differential gene expression was observed in PRA and PRB-rich patient tumors and importantly, PRA-rich gene signatures had poorer survival outcomes. In support of antiprogestin responsiveness of PRA-rich tumors, gene signatures associated with PR antagonists, but not PR agonists, predicted better survival outcomes. The better patient survival associated with PR antagonists versus PR agonists treatments was further reflected in the higher in vivo anti-tumor activity of therapies that combine tamoxifen with PR antagonists and modulators. This study suggests that distinguishing common effects observed due to concomitant interaction of another receptor with its ligand (agonist or antagonist), from unique isoform and ligand-specific effects will inform the development of biomarkers for patient selection and translation of PR-targeted therapies to the clinic.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 21378