Display options
Share it on

Clin Cancer Res. 2021 Sep 15;27(18):4994-5003. doi: 10.1158/1078-0432.CCR-21-1204. Epub 2021 Jul 20.

A Phase I Dose-Escalation and Expansion Study of Telaglenastat in Patients with Advanced or Metastatic Solid Tumors.

Clinical cancer research : an official journal of the American Association for Cancer Research

James J Harding, Melinda Telli, Pamela Munster, Martin H Voss, Jeffrey R Infante, Angela DeMichele, Mark Dunphy, Mai H Le, Chris Molineaux, Keith Orford, Frank Parlati, Sam H Whiting, Mark K Bennett, Nizar M Tannir, Funda Meric-Bernstam

Affiliations

  1. Memorial Sloan Kettering Cancer Center and Weill Medical College, New York, New York. [email protected].
  2. Stanford University School of Medicine, Stanford, California.
  3. University of California at San Francisco, San Francisco, California.
  4. Memorial Sloan Kettering Cancer Center and Weill Medical College, New York, New York.
  5. Sarah Canon Research Institute, Tennessee Oncology PLLC, Nashville, Tennessee.
  6. University of Pennsylvania, Philadelphia, Pennsylvania.
  7. Calithera Biosciences, Inc., South San Francisco, California.
  8. The University of Texas MD Anderson Cancer Center, Houston, Texas.

PMID: 34285061 DOI: 10.1158/1078-0432.CCR-21-1204

Abstract

PURPOSE: Glutamine is a critical fuel for solid tumors. Interference with glutamine metabolism is deleterious to neoplasia in preclinical models. A phase I study of the oral, first-in-class, glutaminase (GLS) inhibitor telaglenastat was conducted in treatment-refractory solid tumor patients to define recommended phase II dose (RP2D) and evaluate safety, pharmacokinetics (PK), pharmacodynamics (PD), and antitumor activity.

PATIENTS AND METHODS: Dose escalation by 3 + 3 design was followed by exploratory tumor-/biomarker-specific cohorts.

RESULTS: Among 120 patients, fatigue (23%) and nausea (19%) were the most common toxicity. Maximum tolerated dose was not reached. Correlative analysis indicated >90% GLS inhibition in platelets at plasma exposures >300 nmol/L, >75% tumoral GLS inhibition, and significant increase in circulating glutamine. RP2D was defined at 800 mg twice-daily. Disease control rate (DCR) was 43% across expansion cohorts (overall response rate 5%, DCR 50% in renal cell carcinoma).

CONCLUSIONS: Telaglenastat is safe, with a favorable PK/PD profile and signal of antitumor activity, supporting further clinical development.

©2021 The Authors; Published by the American Association for Cancer Research.

References

  1. Hensley CT, Wasti AT, DeBerardinis RJ. Glutamine and cancer: cell biology, physiology, and clinical opportunities. J Clin Invest. 2013;123:3678–84. - PubMed
  2. Altman BJ, Stine ZE, Dang CV. From Krebs to clinic: glutamine metabolism to cancer therapy. Nat Rev Cancer. 2016;16:749. - PubMed
  3. DeBerardinis RJ, Mancuso A, Daikhin E, Nissim I, Yudkoff M, Wehrli S, et al. Beyond aerobic glycolysis: transformed cells can engage in glutamine metabolism that exceeds the requirement for protein and nucleotide synthesis. Proc Natl Acad Sci U S A. 2007;104:19345–50. - PubMed
  4. Parlati F, Demo SD, Gross MI, Janes JR, Lewis ER, Mackinnon AL, et al. CB-839, a novel potent and selective glutaminase inhibitor, has broad antiproliferative activity in cell lines derived from both solid tumors and hematological malignancies [abstract]. Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. 2014;p. 74. - PubMed
  5. Cassago A, Ferreira AP, Ferreira IM, Fornezari C, Gomes ER, Greene KS, et al. Mitochondrial localization and structure-based phosphate activation mechanism of Glutaminase C with implications for cancer metabolism. Proc Natl Acad Sci U S A. 2012;109:1092–7. - PubMed
  6. Kung HN, Marks JR, Chi JT. Glutamine synthetase is a genetic determinant of cell type-specific glutamine independence in breast epithelia. PLoS Genet. 2011;7:e1002229. - PubMed
  7. Le A, Lane AN, Hamaker M, Bose S, Gouw A, Barbi J, et al. Glucose-independent glutamine metabolism via TCA cycling for proliferation and survival in B cells. Cell Metab. 2012;15:110–21. - PubMed
  8. Seltzer MJ, Bennett BD, Joshi AD, Gao P, Thomas AG, Ferraris DV, et al. Inhibition of glutaminase preferentially slows growth of glioma cells with mutant IDH1. Cancer Res. 2010;70:8981–7. - PubMed
  9. Son J, Lyssiotis CA, Ying H, Wang X, Hua S, Ligorio M, et al. Glutamine supports pancreatic cancer growth through a KRAS-regulated metabolic pathway. Nature. 2013;496:101–5. - PubMed
  10. van den Heuvel AP, Jing J, Wooster RF, Bachman KE. Analysis of glutamine dependency in non-small cell lung cancer: GLS1 splice variant GAC is essential for cancer cell growth. Cancer Biol Ther. 2012;13:1185–94. - PubMed
  11. Wang JB, Erickson JW, Fuji R, Ramachandran S, Gao P, Dinavahi R, et al. Targeting mitochondrial glutaminase activity inhibits oncogenic transformation. Cancer Cell. 2010;18:207–19. - PubMed
  12. Gameiro PA, Yang J, Metelo AM, Perez-Carro R, Baker R, Wang Z, et al. In vivo HIF-mediated reductive carboxylation is regulated by citrate levels and sensitizes VHL-deficient cells to glutamine deprivation. Cell Metab. 2013;17:372–85. - PubMed
  13. Okazaki A, Gameiro PA, Christodoulou D, Laviollette L, Schneider M, Chaves F, et al. Glutaminase and poly(ADP-ribose) polymerase inhibitors suppress pyrimidine synthesis and VHL-deficient renal cancers. J Clin Invest. 2017;127:1631–45. - PubMed
  14. Commisso C, Davidson SM, Soydaner-Azeloglu RG, Parker SJ, Kamphorst JJ, Hackett S, et al. Macropinocytosis of protein is an amino acid supply route in Ras-transformed cells. Nature. 2013;497:633–7. - PubMed
  15. Alles MC, Gardiner-Garden M, Nott DJ, Wang Y, Foekens JA, Sutherland RL, et al. Meta-analysis and gene set enrichment relative to er status reveal elevated activity of MYC and E2F in the "basal" breast cancer subgroup. PLoS ONE. 2009;4:e4710. - PubMed
  16. Chandriani S, Frengen E, Cowling VH, Pendergrass SA, Perou CM, Whitfield ML, et al. A core MYC gene expression signature is prominent in basal-like breast cancer but only partially overlaps the core serum response. PLoS ONE. 2009;4:e6693. - PubMed
  17. Gatza ML, Lucas JE, Barry WT, Kim JW, Wang Q, Crawford MD, et al. A pathway-based classification of human breast cancer. Proc Natl Acad Sci U S A. 2010;107:6994–9. - PubMed
  18. Gao P, Tchernyshyov I, Chang TC, Lee YS, Kita K, Ochi T, et al. c-Myc suppression of miR-23a/b enhances mitochondrial glutaminase expression and glutamine metabolism. Nature. 2009;458:762–5. - PubMed
  19. Yuneva MO, Fan TW, Allen TD, Higashi RM, Ferraris DV, Tsukamoto T, et al. The metabolic profile of tumors depends on both the responsible genetic lesion and tissue type. Cell Metab. 2012;15:157–70. - PubMed
  20. Collins RRJ, Patel K, Putnam WC, Kapur P, Rakheja D. Oncometabolites: a new paradigm for oncology, metabolism, and the clinical laboratory. Clin Chem. 2017;63:1812–20. - PubMed
  21. Mullen AR, Wheaton WW, Jin ES, Chen PH, Sullivan LB, Cheng T, et al. Reductive carboxylation supports growth in tumour cells with defective mitochondria. Nature. 2011;481:385–8. - PubMed
  22. McBrayer SK, Mayers JR, DiNatale GJ, Shi DD, Khanal J, Chakraborty AA, et al. Transaminase Inhibition by 2-hydroxyglutarate impairs glutamate biosynthesis and redox homeostasis in glioma. Cell. 2018;175:101–16. - PubMed
  23. Gross MI, Demo SD, Dennison JB, Chen L, Chernov-Rogan T, Goyal B, et al. Antitumor activity of the glutaminase inhibitor CB-839 in triple-negative breast cancer. Mol Cancer Ther. 2014;13:890–901. - PubMed
  24. Rodriguez ML, Zhang W, Bennett MK, Emberley E, Gross MI, Janes JR, et al. CB-839, a selective glutaminase inhibitor, synergizes with signal transduction pathway inhibitors to enhance antitumor activity. Cancer Res. 2015;75:15. - PubMed
  25. Parlati F, Bromley-Dulfano S, Demo SD, Janes JR, Gross MI, Lewis ER, et al. Antitumor activity of the glutaminase inhibitor CB-839 in hematological malignances. Blood. 2013;122:4226. - PubMed
  26. Emberley E, Bennett MK, Chen J, Gross MI, Huang T, Li W, et al. CB-839, a selective glutaminase inhibitor, has antitumor activity in renal cell carcinoma and synergizes with everolimus and receptor tyrosine kinase inhibitors. Eur J Cancer. 2016;69:S124. - PubMed
  27. Mackinnon AL, Chen J, Gross MI, Marguier G, Shwonek PJ, Sotirovska N, et al. Targeting tumor glutamine metabolism with CB-839 enhances the efficacy of immune checkpoint inhibitors. J Immunother Cancer. 2016;4:P224. - PubMed
  28. Parlati F, Chernov-Rogan T, Demo SD, Gross MI, Janes JR, Kawas R, et al. Antitumor activity of novel, potent, selective, and orally-bioavailable glutaminase inhibitors. Cancer Res. 2013;73:8. - PubMed
  29. Parlati F, Gross MI, Janes JR, Lewis ER, Mackinnon AL, Rodriguez ML, et al. Glutaminase inhibitor CB-839 synergizes with pomalidomide in preclinical multiple myeloma models. Blood. 2014;124:4720. - PubMed
  30. Thompson RM, Dytfeld D, Reyes L, Robinson RM, Smith B, Manevich Y, et al. Glutaminase inhibitor CB-839 synergizes with carfilzomib in resistant multiple myeloma cells. Oncotarget. 2017;8:35863–76. - PubMed
  31. Lampa M, Arlt H, He T, Ospina B, Reeves J, Zhang B, et al. Glutaminase is essential for the growth of triple-negative breast cancer cells with a deregulated glutamine metabolism pathway and its suppression synergizes with mTOR inhibition. PLoS ONE. 2017;12:e0185092. - PubMed
  32. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45:228–47. - PubMed
  33. Byrne MJ, Nowak AK. Modified RECIST criteria for assessment of response in malignant pleural mesothelioma. Ann Oncol. 2004;15:257–60. - PubMed
  34. Lowery MA, Burris HAJanku F, Shroff RT, Cleary JM, Azad NS, et al. Safety and activity of ivosidenib in patients with IDH1-mutant advanced cholangiocarcinoma: a phase 1 study. Lancet Gastroenterol Hepatol. 2019;4:711–20. - PubMed
  35. Romero R, Sayin VI, Davidson SM, Bauer MR, Singh SX, LeBoeuf SE, et al. Keap1 loss promotes Kras-driven lung cancer and results in dependence on glutaminolysis. Nat Med. 2017;23:1362–8. - PubMed
  36. Motzer R, Lee C-H, Emamekhoo H, Matrana M, Percent I, Hsieh J, et al. ENTRATA: randomized, double-blind, phase 2 study of telaglenastat (tela; CB-839) + everolimus (E) vs. placebo (pbo) + E in patients (pts) with advanced/metastatic renal cell carcinoma (mRCC). Ann Oncol. 2019;30:v851–934. - PubMed
  37. Tannir N, Agarwal N, Porta C, Lawrence N, Motzer R, Lee R, et al. CANTATA: Primary analysis of a global, randomized, placebo (Pbo)-controlled, double-blind trial of telaglenastat (CB-839) + cabozantinib vs. Pbo + cabozantinib in patients (pts) with advanced/metastatic renal cell carcinoma (mRCC) that progressed on immune checkpoint inhibitor (ICI) or anti-angiogenic therapies. J Clin Oncol. 2021;39:15. - PubMed
  38. KEAPSAKE: a study of Telaglenastat (CB-839) with standard-of-care chemoimmunotherapy in 1L KEAP1/NRF2-mutated, nonsquamous NSCLC (KEAPSAKE). - PubMed

Publication Types