1. Academic Validation
  2. The Dysregulated Pharmacology of Clinically Relevant ESR1 Mutants is Normalized by Ligand-activated WT Receptor

The Dysregulated Pharmacology of Clinically Relevant ESR1 Mutants is Normalized by Ligand-activated WT Receptor

  • Mol Cancer Ther. 2020 Jul;19(7):1395-1405. doi: 10.1158/1535-7163.MCT-19-1148.
Kaitlyn J Andreano 1 Jennifer G Baker 1 Sunghee Park 1 Rachid Safi 1 Sandeep Artham 1 Steffi Oesterreich 2 3 Rinath Jeselsohn 4 Myles Brown 4 Sarah Sammons 5 Suzanne E Wardell 1 Ching-Yi Chang 1 John D Norris 1 Donald P McDonnell 6
Affiliations

Affiliations

  • 1 Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina.
  • 2 Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.
  • 3 Womens Cancer Research Center, University of Pittsburgh Cancer Institute and Magee-Women Research Institute, Pittsburgh, Pennsylvania.
  • 4 Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
  • 5 Department of Medical Oncology, Duke Cancer Institute, Duke University, Durham, North Carolina.
  • 6 Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina. [email protected].
Abstract

The Estrogen Receptor (ER/ESR1) is expressed in a majority of breast cancers and drugs that inhibit ER signaling are the cornerstone of breast Cancer pharmacotherapy. Currently, Aromatase inhibitors are the frontline endocrine interventions of choice although their durability in metastatic disease is limited by activating point mutations within the ligand-binding domain of ESR1 that permit ligand-independent activation of the receptor. It has been suggested that the most commonly occurring ESR1 mutations would likely compromise the clinical activity of selective Estrogen Receptor downregulators and selective Estrogen Receptor modulators (SERMs) when used as second-line therapies. It was unclear, however, how these mutations, which are likely coexpressed in cells with ERWT, may impact response to ER ligands in a clinically meaningful manner. To address this issue, we dissected the molecular mechanism(s) underlying ESR1-mutant pharmacology in models relevant to metastatic disease. These studies revealed that the response of ESR1 mutations to ligands was dictated primarily by the relative coexpression of ERWT in cells. Specifically, dysregulated pharmacology was only evident in cells in which the mutants were overexpressed relative to ligand-activated ERWT; a finding that highlights the role of allelism in determining ER-mutant pharmacology. Importantly, we demonstrated that the antagonist activity of the SERM, lasofoxifene, was not impacted by mutant status; a finding that has led to its clinical evaluation as a treatment for patients with advanced ER-positive breast Cancer whose tumors harbor ESR1 mutations.

Figures
Products