Oncotarget

Research Papers:

Different metabolic responses to PI3K inhibition in NSCLC cells harboring wild-type and G12C mutant KRAS

Elisa Caiola, Laura Brunelli, Mirko Marabese, Massimo Broggini _, Monica Lupi and Roberta Pastorelli

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2016; 7:51462-51472. https://doi.org/10.18632/oncotarget.9849

Metrics: PDF 1569 views  |   HTML 2418 views  |   ?  


Abstract

Elisa Caiola1,*, Laura Brunelli2,*, Mirko Marabese1, Massimo Broggini1, Monica Lupi3, Roberta Pastorelli2

1Laboratory of Molecular Pharmacology, Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche “Mario Negri”, Milan, Italy

2Protein and Gene Biomarkers Unit, Laboratory of Mass Spectrometry, Department of Environmental Health Sciences, IRCCS-Istituto di Ricerche Farmacologiche “Mario Negri”, Milan, Italy

3Laboratory of Cancer Pharmacology, Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche “Mario Negri”, Milan, Italy

*These authors have contributed equally to this work

Correspondence to:

Massimo Broggini, email: [email protected]

Roberta Pastorelli, email: [email protected]

Keywords: KRAS, NSCLC, metabolomics, BEZ235, BKM120

Received: November 24, 2015     Accepted: May 22, 2016     Published: June 06, 2016

ABSTRACT

KRAS mutations in non-small-cell lung cancer (NSCLC) patients are considered a negative predictive factor and indicate poor response to anticancer treatments. KRAS mutations lead to activation of the PI3K/akt/mTOR pathway, whose inhibition remains a challenging clinical target. Since the PI3K/akt/mTOR pathway and KRAS oncogene mutations all have roles in cancer cell metabolism, we investigated whether the activity of PI3K/akt/mTOR inhibitors (BEZ235 and BKM120) in cells harboring different KRAS status is related to their metabolic effect. Isogenic NSCLC cell clones expressing wild-type (WT) and mutated (G12C) KRAS were used to determine the response to BEZ235 and BKM120. Metabolomics analysis indicated the impairment of glutamine in KRAS-G12C and serine metabolism in KRAS-WT, after pharmacological blockade of the PI3K signaling, although the net effect on cell growth, cell cycle distribution and caspase activation was similar. PI3K inhibitors caused autophagy in KRAS-WT, but not in KRAS-G12C, where there was a striking decrease in ammonia production, probably a consequence of glutamine metabolism impairment.

These findings lay the grounds for more effective therapeutic combinations possibly distinguishing wild-type and mutated KRAS cancer cells in NSCLC, exploiting their different metabolic responses to PI3K/akt/mTOR inhibitors.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 9849