Multiple Components of Protein Homeostasis Pathway Can Be Targeted to Produce Drug Synergies with VCP Inhibitors in Ovarian Cancer

CANCERS(2022)

Cited 4|Views18
No score
Abstract
Simple Summary The increased dependency of cancer cells on protein homeostasis creates a vulnerability that can be exploited using protein homeostasis inhibiting drugs. Recently, valosin-containing protein has emerged as a key component of the pathway and a novel therapeutic target for cancer treatment. The aim of our research is to explore the potential synergies between VCP inhibitors and other agents that disrupt protein homeostasis and to provide mechanistic understanding of drug synergies targeting multiple components of protein homeostasis. Our results indicate that VCP inhibitors can be combined with other endoplasmic reticulum stress-inducing agents to produce synergistic cytotoxicity in ovarian cancer cells. Protein quality control mechanisms play an important role in cancer progression by providing adaptive responses and morphologic stability against genome-wide copy number alterations, aneuploidy, and conformation-altering somatic mutations. This dependency on protein quality control mechanisms creates a vulnerability that may be exploited for therapeutic benefits by targeting components of the protein quality control mechanism. Recently, valosin-containing protein (VCP), also known at p97 AAA-ATPase, has emerged as a druggable target in cancer cells to affect their dependency on protein quality control. Here, we show that VCP inhibitors induce cytotoxicity in several ovarian cancer cell lines and these compounds act synergistically with mifepristone, a drug previously shown to induce an atypical unfolded protein response. Although mifepristone at a clinically achievable dose induces a weak unfolded protein response, it enhances the cytotoxic effects of VCP inhibitor CB-5083. Mechanistically, mifepristone blocks the cytoprotective effect of ATF6 in response to endoplasmic reticulum (ER) stress while activating the cytotoxic effects of ATF4 and CHOP through the HRI (EIF2AK1)-mediated signal transduction pathway. In contrast, CB-5083 activates ATF4 and CHOP through the PERK (EIF2AK3)-mediated signaling pathway. This combination activates ATF4 and CHOP while blocking the adaptive response provided by ATF6, resulting in increased cytotoxic effects and synergistic drug interaction.
More
Translated text
Key words
VCP inhibitors,mifepristone,ovarian cancer,proteostasis,cancer therapy
AI Read Science
Must-Reading Tree
Example
Generate MRT to find the research sequence of this paper
Chat Paper
Summary is being generated by the instructions you defined