Abstract

Abstract There have been tremendous strides in the development of therapies for the treatment of NSCLC over the past decade. Nevertheless, the most common recurrent mutation driving the growth of NSCLC, mutant KRAS, accounting for ~25% of patients with advanced NSCLC, remains without an effective therapy. Thus, novel therapies are critically needed to improve the lives of patients suffering from KRAS-driven lung cancers. Protein phosphatase 2A (PP2A) is a serine threonine-directed tumor-suppressive phosphatase that is dysregulated and deactivated in cancer by multiple mechanisms including somatic mutation, suppression of individual subunits, increased expression of endogenous PP2A inhibitors and changes in post-translational modifications to the catalytic C subunit. Given its central role in regulating many key cellular processes and its role in human disease pathogenesis, many efforts have been developed to therapeutically target PP2A. Our group has developed first-in-class direct small-molecule activators of PP2A (SMAPs) by using the chemical scaffold of the FDA-approved tricyclic neuroleptics. We have used molecular modeling, hydroxyl radical footprinting and cryo-electron microscopy to structurally resolve the mechanism of action of SMAP-mediated growth suppression. Specifically, SMAPs protect the regulatory c-terminal tail of the catalytic subunit through changes in the methylation of the terminal leucine (L309) that induces the holoenzyme heterotrimerization and increased substrate-directed catalysis. SMAP treatment of KRAS-mutant lung cancer cells induced apoptosis and decreased the phosphorylation of PP2A targets including AKT and ERK. We found that treatment of xenograft, PDX, and GEMM mouse models of KRAS-mutant lung cancer cells with SMAPs resulted in marked tumor growth inhibition. Taken together, these studies demonstrate the potential of using SMAPs as an approach to treat KRAS-mutant NSCLC. We also established that PP2A activity defines the response of KRAS-mutant lung cancer cells across library of over 200 kinase inhibitors. In vivo studies using xenograft mouse models of KRAS-mutant lung adenocarcinoma demonstrated that SMAP in combination with MEK inhibitor resulted in tumor regression. Our studies have identified a combination of drugs that is effective in vivo for the treatment of KRAS-mutant lung cancer. Our strategy of reactivating endogenous PP2A may be applicable to the treatment of other KRAS-driven cancers and represents a first step into a new territory of developing small-molecule activators of tumor-suppressor proteins. Citation Format: Jaya Sangodkar, Jukka Westermarck, Derek Taylor, Goutham Narla. Therapeutic reactivation of the protein phosphatase 2A (PP2A) for the treatment of KRAS-driven cancers [abstract]. In: Proceedings of the AACR Special Conference on Targeting RAS-Driven Cancers; 2018 Dec 9-12; San Diego, CA. Philadelphia (PA): AACR; Mol Cancer Res 2020;18(5_Suppl):Abstract nr A48.

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call