Abstract

Abstract During tumorigenesis cancer cells continuously encountermetabolic bottlenecks as a result of accelerated growth, overall increased metabolic demand and increased oxidative stress due to the formation of reactive oxygen species. We use a combination of genetically-engineered mouse models, an accelerated CRISPR/Cas9-based experimentalplatformand biochemical approaches to identify metabolic liabilities that can be exploited using novel targeted therapies.Lung cancer, the leading cause of cancer-related deaths worldwide,is the most common cancer type to acquire mutually exclusive gain-of-function mutations in the anti-oxidant transcription factor NRF2or loss-of-function mutations in its negative regulator KEAP1. Loss of Keap1activates Nrf2, increases antioxidant production and dramatically accelerates KRAS-driven lung cancer. Our data are in line with mounting evidence demonstrating that antioxidants promote cancer progression. Our studies focus on elucidating the mechanisms underlying novel metabolic dependencies in KEAP1mutant tumors and explore the therapeutic potential of targeting metabolism in highly relevant pre-clinical mouse and human lung cancer models. We observe that the ability of KEAP1mutant tumors to divert their metabolism towards antioxidant production comes with a cost, generating multiple metabolic vulnerabilities, including a dependency on glutamine metabolism that can be therapeutically exploited using DRP-104, a novel broad acting glutamine antagonist which inhibits all glutamine-consuming reactions. In addition, we have extended our findings to demonstrate that cancers with high antioxidant capacity exhibit a general dependency on exogenous non-essential amino acids (NEAAs) that is driven by the Nrf2-dependent secretion of glutamate through system xc-(XCT), which limits intracellular glutamate pools that are required for NEAA synthesis. This dependency can be therapeutically targeted by dietary restriction or enzymatic depletion of individual NEAAs. Importantly, limiting endogenous glutamate levels by glutaminase inhibition can sensitize tumors without alterations in Keap1/Nrf2 pathway to dietary restriction of NEAAs. Our findings identify a metabolic strategy to therapeutically target cancers with genetic or pharmacologic activation of the Nrf2 antioxidant response pathway by restricting exogenous sources of NEAAs. Citation Format: Sarah E. LeBoeuf, Warren Wu, Triantafyllia Karakousi, Robert Wild, Thales Papagiannakopoulos. Uncovering metabolic bottlenecks in KEAP1 mutant lung cancer [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 2569.

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