Oncotarget

Research Papers:

This article has been corrected. Correction in: Oncotarget. 2018; 9:33866.

Drug-Targeted Inhibition of Peroxisome Proliferator-Activated Receptorγ Enhances the Chemopreventive Effect of Anti-Estrogen

Hongyan Yuan, Levy Kopelovich, Yuzhi Yin, Jin Lu and Robert I Glazer _

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2012; 3:345-358. https://doi.org/10.18632/oncotarget.457

Metrics: PDF 3266 views  |   HTML 3357 views  |   ?  


Abstract

Hongyan Yuan1, Levy Kopelovich2, Yuzhi Yin1,3, Jin Lu1 and Robert I. Glazer1

1 Department of Oncology, Georgetown University School of Medicine, and Lombardi Comprehensive Cancer Center, Washington, DC

2 Chemoprevention Branch, National Cancer Institute, Bethesda, MD

3 Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD

Received: February 20, 2012; Accepted: April 9, 2012; Published: April 13, 2012;

Keywords: PPARγ, ERα, fulvestrant, GW9662

Correspondence:

Robert I. Glazer, email:

Abstract

The peroxisome proliferator-activated receptorγ (PPARγ) is a key regulator of metabolism, proliferation, inflammation and differentiation, and upregulates tumor suppressor genes, such as PTEN, BRCA1 and PPARγ itself. Examination of mammary carcinogenesis in transgenic mice expressing the dominant-negative Pax8PPARγ fusion protein revealed that tumors were estrogen receptorα (ER)-positive and sensitive to the ER antagonist, fulvestrant. Here we evaluated whether administration of an irreversible PPARγ inhibitor in vivo could similarly induce ER expression in otherwise ER-negative mammary tumors following induction of carcinogenesis, and sensitize them to the antitumor effects of fulvestrant. In addition, we wished to determine whether the effect of GW9662 was associated with a PPAR-selective gene expression profile. Mammary carcinogenesis was induced in wild-type FVB mice by treatment with medroxyprogesterone and dimethylbenz(a)anthracene (DMBA) that were subsequently maintained on a diet supplemented with 0.1% GW9662, and tumorigenesis and gene expression profiling of the resulting tumors were determined. Administration of GW9962 resulted in ER+ tumors that were highly sensitive to fulvestrant. Tumors from GW9662-treated animals exhibited reduced expression of a metabolic gene profile indicative of PPARγ inhibition, including PPARγ itself. Additionally, GW9662 upregulated the expression of several genes associated with the transcription, processing, splicing and translation of RNA. This study is the first to show that an irreversible PPARγ inhibitor can mimic a dominant-negative PPARγ transgene to elicit the development of ER-responsive tumors. These findings suggest that it may be possible to pharmacologically influence the responsiveness of tumors to anti-estrogen therapy.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 457