Your browser doesn't support javascript.
loading
Identifying strategies to target the metabolic flexibility of tumours.
Méndez-Lucas, Andrés; Lin, Wei; Driscoll, Paul C; Legrave, Nathalie; Novellasdemunt, Laura; Xie, Chencheng; Charles, Mark; Wilson, Zena; Jones, Neil P; Rayport, Stephen; Rodríguez-Justo, Manuel; Li, Vivian; MacRae, James I; Hay, Nissim; Chen, Xin; Yuneva, Mariia.
Affiliation
  • Méndez-Lucas A; The Francis Crick Institute, London, UK.
  • Lin W; The Francis Crick Institute, London, UK.
  • Driscoll PC; The Francis Crick Institute, London, UK.
  • Legrave N; The Francis Crick Institute, London, UK.
  • Novellasdemunt L; The Francis Crick Institute, London, UK.
  • Xie C; Department of Internal Medicine, University of South Dakota, Sanford School of Medicine, Vermillion, SD, USA.
  • Charles M; Cancer Research UK, Therapeutic Discovery Laboratories, Cambridge, UK.
  • Wilson Z; Bioscience, Discovery, Oncology R&D, AstraZeneca, Macclesfield, UK.
  • Jones NP; Cancer Research UK, Therapeutic Discovery Laboratories, Cambridge, UK.
  • Rayport S; Department of Psychiatry, Columbia University, New York, NY, USA.
  • Rodríguez-Justo M; Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA.
  • Li V; Histopathology Department, University College London, London, UK.
  • MacRae JI; The Francis Crick Institute, London, UK.
  • Hay N; The Francis Crick Institute, London, UK.
  • Chen X; Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.
  • Yuneva M; Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA, USA.
Nat Metab ; 2(4): 335-350, 2020 04.
Article in En | MEDLINE | ID: mdl-32694609
ABSTRACT
Plasticity of cancer metabolism can be a major obstacle to efficient targeting of tumour-specific metabolic vulnerabilities. Here, we identify the compensatory mechanisms following the inhibition of major pathways of central carbon metabolism in c-MYC-induced liver tumours. We find that, while inhibition of both glutaminase isoforms (Gls1 and Gls2) in tumours considerably delays tumourigenesis, glutamine catabolism continues, owing to the action of amidotransferases. Synergistic inhibition of both glutaminases and compensatory amidotransferases is required to block glutamine catabolism and proliferation of mouse and human tumour cells in vitro and in vivo. Gls1 deletion is also compensated for by glycolysis. Thus, co-inhibition of Gls1 and hexokinase 2 significantly affects Krebs cycle activity and tumour formation. Finally, the inhibition of biosynthesis of either serine (Psat1-KO) or fatty acid (Fasn-KO) is compensated for by uptake of circulating nutrients, and dietary restriction of both serine and glycine or fatty acids synergistically suppresses tumourigenesis. These results highlight the high flexibility of tumour metabolism and demonstrate that either pharmacological or dietary targeting of metabolic compensatory mechanisms can improve therapeutic outcomes.
Subject(s)

Full text: 1 Database: MEDLINE Main subject: Liver Neoplasms Limits: Animals / Humans Language: En Year: 2020 Type: Article

Full text: 1 Database: MEDLINE Main subject: Liver Neoplasms Limits: Animals / Humans Language: En Year: 2020 Type: Article