Autophagy is required for crizotinib-induced apoptosis in MET-amplified gastric cancer cells

Rebecca D. Schroeder, Woonyoung Choi, David S. Hong, David J. McConkey

Research output: Contribution to journalArticlepeer-review

7 Scopus citations

Abstract

MET amplification has been clinically credentialed as a therapeutic target in gastric cancer, but the molecular mechanisms underlying sensitivity and resistance to MET inhibitors are still not well understood. Using whole-genome mRNA expression profiling, we identified autophagy as a top molecular pathway that was activated by the MET inhibitor crizotinib in drug-sensitive human gastric cancer cells, and functional studies confirmed that crizotinib increased autophagy levels in the drugsensitive cells in a concentration-dependent manner. We then used chemical and molecular approaches to inhibit autophagy in order to define its role in cell death. The clinically available inhibitor of autophagy, chloroquine, or RNAi-mediated knockdown of two obligate components of the autophagy pathway (ATG5 and ATG7) blocked cell death induced by crizotinib or RNAi-mediated knockdown of MET, and mechanistic studies localized the effects of autophagy to cytochrome c release from the mitochondria. Overall, the data reveal a novel relationship between autophagy and apoptosis in gastric cancer cells exposed to MET inhibitors. The observations suggest that autophagy inhibitors should not be used to enhance the effects of MET inhibitors in gastric cancer patients.

Original languageEnglish (US)
Pages (from-to)51675-51687
Number of pages13
JournalOncotarget
Volume8
Issue number31
DOIs
StatePublished - Feb 7 2017

Keywords

  • Autophagy
  • Gastric cancer
  • MET amplification
  • Resistance mechanism

ASJC Scopus subject areas

  • Oncology

Fingerprint

Dive into the research topics of 'Autophagy is required for crizotinib-induced apoptosis in MET-amplified gastric cancer cells'. Together they form a unique fingerprint.

Cite this