Original Research
Pharmacologic Activation of the G Protein–Coupled Estrogen Receptor Inhibits Pancreatic Ductal Adenocarcinoma

https://doi.org/10.1016/j.jcmgh.2020.04.016Get rights and content
Under a Creative Commons license
open access

Background & Aims

Female sex is associated with lower incidence and improved clinical outcomes for most cancer types including pancreatic ductal adenocarcinoma (PDAC). The mechanistic basis for this sex difference is unknown. We hypothesized that estrogen signaling may be responsible, despite the fact that PDAC lacks classic nuclear estrogen receptors.

Methods

Here we used murine syngeneic tumor models and human xenografts to determine that signaling through the nonclassic estrogen receptor G protein-coupled estrogen receptor (GPER) on tumor cells inhibits PDAC.

Results

Activation of GPER with the specific, small molecule, synthetic agonist G-1 inhibited PDAC proliferation, depleted c-Myc and programmed death ligand 1 (PD-L1), and increased tumor cell immunogenicity. Systemically administered G-1 was well-tolerated in PDAC bearing mice, induced tumor regression, significantly prolonged survival, and markedly increased the efficacy of PD-1 targeted immune therapy. We detected GPER protein in a majority of spontaneous human PDAC tumors, independent of tumor stage.

Conclusions

These data, coupled with the wide tissue distribution of GPER and our previous work showing that G-1 inhibits melanoma, suggest that GPER agonists may be useful against a range of cancers that are not classically considered sex hormone responsive and that arise in tissues outside of the reproductive system.

Keywords

Pancreatic Ductal Adenocarcinoma
PDAC
G Protein-Coupled Estrogen Receptor
GPER

Abbreviations used in this paper

DMEM
Dulbecco modified Eagle medium
FBS
fetal bovine serum
GPER
G protein-coupled estrogen receptor
HPDE
human pancreatic ductal epithelial
PBS
phosphate-buffered saline
PDAC
pancreatic ductal adenocarcinoma
PD-L1
programmed death ligand 1

Cited by (0)

Conflicts of interest These authors disclose the following: C.A.N. is listed as an inventor on a patent application held by the University of Pennsylvania related to this work (PCT/US2017/035278) and is a cofounder and current employee of the Penn Center for Innovation supported startup Linnaeus Therapeutics Inc. T.W.R. is listed as an inventor on a patent application (PCT/US2017/035278) held by the University of Pennsylvania related to this work and is a cofounder and consultant to the Penn Center for Innovation supported startup Linnaeus Therapeutics Inc. The remaining authors disclose no conflicts.

Funding T.W.R. is supported by a grant from the NIH/NCI (R01 CA163566), a Penn/Wistar Institute N.I.H. SPORE (P50CA174523), the Melanoma Research Foundation, and the Dermatology Foundation. C.A.N was supported by an NIH/NIAMS training grant (T32 AR0007465-32) and an NIH/NCI F31 NRSA Individual Fellowship (F31 CA206325). This work was supported in part by the Penn Skin Biology and Diseases Resource-based Center (P30-AR069589). This work was also supported in part by a phase I STTR from the NIH/NCI (R41CA228695). The contents are solely the responsibility of the authors and do not necessarily represent the official views of the NIH.