MET Inhibition Elicits PGC1α-Dependent Metabolic Reprogramming in Glioblastoma

Cancer Res. 2020 Jan 1;80(1):30-43. doi: 10.1158/0008-5472.CAN-19-1389. Epub 2019 Nov 6.

Abstract

The receptor kinase c-MET has emerged as a target for glioblastoma therapy. However, treatment resistance emerges inevitably. Here, we performed global metabolite screening with metabolite set enrichment coupled with transcriptome and gene set enrichment analysis and proteomic screening, and identified substantial reprogramming of tumor metabolism involving oxidative phosphorylation and fatty acid oxidation (FAO) with substantial accumulation of acyl-carnitines accompanied by an increase of PGC1α in response to genetic (shRNA and CRISPR/Cas9) and pharmacologic (crizotinib) inhibition of c-MET. Extracellular flux and carbon tracing analyses (U-13C-glucose, U-13C-glutamine, and U-13C-palmitic acid) demonstrated enhanced oxidative metabolism, which was driven by FAO and supported by increased anaplerosis of glucose carbons. These findings were observed in concert with increased number and fusion of mitochondria and production of reactive oxygen species. Genetic interference with PGC1α rescued this oxidative phenotype driven by c-MET inhibition. Silencing and chromatin immunoprecipitation experiments demonstrated that cAMP response elements binding protein regulates the expression of PGC1α in the context of c-MET inhibition. Interference with both oxidative phosphorylation (metformin, oligomycin) and β-oxidation of fatty acids (etomoxir) enhanced the antitumor efficacy of c-MET inhibition. Synergistic cell death was observed with c-MET inhibition and gamitrinib treatment. In patient-derived xenograft models, combination treatments of crizotinib and etomoxir, and crizotinib and gamitrinib were significantly more efficacious than single treatments and did not induce toxicity. Collectively, we have unraveled the mechanistic underpinnings of c-MET inhibition and identified novel combination therapies that may enhance its therapeutic efficacy. SIGNIFICANCE: c-MET inhibition causes profound metabolic reprogramming that can be targeted by drug combination therapies.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Antineoplastic Combined Chemotherapy Protocols / pharmacology*
  • Antineoplastic Combined Chemotherapy Protocols / therapeutic use
  • Brain Neoplasms / drug therapy*
  • Brain Neoplasms / metabolism
  • Brain Neoplasms / pathology
  • Carnitine / analogs & derivatives
  • Carnitine / metabolism
  • Cell Line, Tumor
  • Cell Proliferation / drug effects
  • Cell Respiration / drug effects
  • Crizotinib / pharmacology
  • Crizotinib / therapeutic use
  • Drug Synergism
  • Epoxy Compounds / pharmacology
  • Epoxy Compounds / therapeutic use
  • Fatty Acids / metabolism
  • Gene Expression Profiling
  • Glioblastoma / drug therapy*
  • Glioblastoma / genetics
  • Glioblastoma / metabolism
  • Glioblastoma / pathology
  • Glycolysis / drug effects
  • Guanidines / pharmacology
  • Guanidines / therapeutic use
  • Humans
  • Lactams, Macrocyclic / pharmacology
  • Lactams, Macrocyclic / therapeutic use
  • Metabolomics
  • Mice
  • Mitochondria / metabolism
  • Mitochondrial Dynamics / drug effects
  • Oxidative Phosphorylation / drug effects
  • Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha / metabolism*
  • Proteomics
  • Proto-Oncogene Proteins c-met / antagonists & inhibitors*
  • Proto-Oncogene Proteins c-met / genetics
  • Proto-Oncogene Proteins c-met / metabolism
  • Reactive Oxygen Species / metabolism
  • Xenograft Model Antitumor Assays

Substances

  • Epoxy Compounds
  • Fatty Acids
  • Guanidines
  • Lactams, Macrocyclic
  • PPARGC1A protein, human
  • Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha
  • Reactive Oxygen Species
  • acylcarnitine
  • gamitrinib-G4
  • Crizotinib
  • MET protein, human
  • Proto-Oncogene Proteins c-met
  • etomoxir
  • Carnitine