Your browser doesn't support javascript.
loading
Targeting glutamine metabolism sensitizes pancreatic cancer to PARP-driven metabolic catastrophe induced by ß-lapachone.
Chakrabarti, Gaurab; Moore, Zachary R; Luo, Xiuquan; Ilcheva, Mariya; Ali, Aktar; Padanad, Mahesh; Zhou, Yunyun; Xie, Yang; Burma, Sandeep; Scaglioni, Pier P; Cantley, Lewis C; DeBerardinis, Ralph J; Kimmelman, Alec C; Lyssiotis, Costas A; Boothman, David A.
Afiliación
  • Chakrabarti G; Department of Pharmacology, University of Texas Southwestern Medical Center, 6001 Forest Park Drive, Dallas, 75390-8807 TX USA ; Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX USA.
  • Moore ZR; Department of Pharmacology, University of Texas Southwestern Medical Center, 6001 Forest Park Drive, Dallas, 75390-8807 TX USA ; Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX USA.
  • Luo X; Department of Pharmacology, University of Texas Southwestern Medical Center, 6001 Forest Park Drive, Dallas, 75390-8807 TX USA ; Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX USA.
  • Ilcheva M; Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX USA.
  • Ali A; Touchstone Diabetes Center, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX USA.
  • Padanad M; Department of Internal Medicine, Weill Cornell Medical College, 413 East 69th Street, BB-1362, New York, NY 10021 USA.
  • Zhou Y; Department of Bioinformatics and Biostatistics, Clinical Sciences, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390 USA.
  • Xie Y; Department of Bioinformatics and Biostatistics, Clinical Sciences, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390 USA.
  • Burma S; Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX USA.
  • Scaglioni PP; Department of Internal Medicine, Weill Cornell Medical College, 413 East 69th Street, BB-1362, New York, NY 10021 USA.
  • Cantley LC; Department of Medicine, Weill Cornell Medical College, 413 East 69th Street, BB-1362, New York, NY 10021 USA.
  • DeBerardinis RJ; Children's Medical Center Research Institute, UT Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390 USA.
  • Kimmelman AC; Department of Radiation Oncology, Division of Genomic Stability and DNA Repair, Dana-Farber Cancer Institute, Boston, MA 02215 USA.
  • Lyssiotis CA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109 USA ; Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI 48109 USA.
  • Boothman DA; Department of Pharmacology, University of Texas Southwestern Medical Center, 6001 Forest Park Drive, Dallas, 75390-8807 TX USA ; Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX USA.
Cancer Metab ; 3: 12, 2015.
Article en En | MEDLINE | ID: mdl-26462257
ABSTRACT

BACKGROUND:

Pancreatic ductal adenocarcinomas (PDA) activate a glutamine-dependent pathway of cytosolic nicotinamide adenine dinucleotide phosphate (NADPH) production to maintain redox homeostasis and support proliferation. Enzymes involved in this pathway (GLS1 (mitochondrial glutaminase 1), GOT1 (cytoplasmic glutamate oxaloacetate transaminase 1), and GOT2 (mitochondrial glutamate oxaloacetate transaminase 2)) are highly upregulated in PDA, and among these, inhibitors of GLS1 were recently deployed in clinical trials to target anabolic glutamine metabolism. However, single-agent inhibition of this pathway is cytostatic and unlikely to provide durable benefit in controlling advanced disease.

RESULTS:

Here, we report that reducing NADPH pools by genetically or pharmacologically (bis-2-(5-phenylacetamido-1,2,4-thiadiazol-2-yl)ethyl sulfide (BPTES) or CB-839) inhibiting glutamine metabolism in mutant Kirsten rat sarcoma viral oncogene homolog (KRAS) PDA sensitizes cell lines and tumors to ß-lapachone (ß-lap, clinical form ARQ761). ß-Lap is an NADPHquinone oxidoreductase (NQO1)-bioactivatable drug that leads to NADPH depletion through high levels of reactive oxygen species (ROS) from the futile redox cycling of the drug and subsequently nicotinamide adenine dinucleotide (NAD)+ depletion through poly(ADP ribose) polymerase (PARP) hyperactivation. NQO1 expression is highly activated by mutant KRAS signaling. As such, ß-lap treatment concurrent with inhibition of glutamine metabolism in mutant KRAS, NQO1 overexpressing PDA leads to massive redox imbalance, extensive DNA damage, rapid PARP-mediated NAD+ consumption, and PDA cell death-features not observed in NQO1-low, wild-type KRAS expressing cells.

CONCLUSIONS:

This treatment strategy illustrates proof of principle that simultaneously decreasing glutamine metabolism-dependent tumor anti-oxidant defenses and inducing supra-physiological ROS formation are tumoricidal and that this rationally designed combination strategy lowers the required doses of both agents in vitro and in vivo. The non-overlapping specificities of GLS1 inhibitors and ß-lap for PDA tumors afford high tumor selectivity, while sparing normal tissue.
Palabras clave

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Idioma: En Revista: Cancer Metab Año: 2015 Tipo del documento: Article

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Idioma: En Revista: Cancer Metab Año: 2015 Tipo del documento: Article