778
Views
0
CrossRef citations to date
0
Altmetric
Review

Glutaminase inhibitors: a patent review

, , &
Pages 823-835 | Received 28 Jul 2018, Accepted 27 Sep 2018, Published online: 11 Oct 2018

References

  • Rj D, Sayed N, Ditsworth D, et al. Brick by brick: metabolism and tumor cell growth. Gurr Opin Genet Dev. 2018;18:54–61.
  • Kroemer G, Pouyssegur J. Tumor cell metabolism: cancer’s Achilles’ heel. Cancer Cell. 2008;13:472–482.
  • DeBerardinis RJ, Mancuso A, Daikhin E, 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 USA. 2007;104:19345–19350.
  • Katt WP, Ramachandran S, Erickson JW, et al. Dibenzophenanthridines as inhibitors of glutaminase C and cancer cell proliferation. Mol Cancer Ther. 2012;11(6):1269–1278.
  • Medina MA. Glutamine and Cancer. J Nutr. 2001;131(9 Suppl):2539S–2542S.
  • Deberardinis RJ, Lum JJ, Hatzivassiliou G, et al. The biology of cancer: metabolic reprogramming fuels cell growth and proliferation. Cell Metab. 2008;7:11–20.
  • Hensley CT, Wasti AT, Deberardinis RJ. Glutamine and Cancer: cell Biology, Physiology, and Clinical Opportunities. J Clin Invest. 2013;123:3678–3684.
  • DeBerardinis RJ, Cheng T. Q’s next: the diverse functions of glutamine in metabolism, cell biology and cancer. Oncogene. 2010;29(3):313–324.
  • Hu W, Zhang C, Wu R, et al. Glutaminase 2, a novel p53 target gene regulating energy metabolism and antioxidant function. Proc Natl Acad Sci USA. 2010;107:7455–7460.
  • Eagle H, Oyama VI, Levy M, et al. The growth response of mammalian cells in tissue culture to L-glutamine and L-glutamic acid. J Biol Chem. 1956;218:607–616.
  • Eagle H. Nutrition needs of mammalian cells in tissue culture. Science. 1955;122:501–514.
  • Kovacevic Z, Morris HP. The role of glutamine in the oxidative metabolism of malignant cells. Cancer Res. 1972;32:326–333.
  • Lavietes BB, Regan DH, Demopoulos HB. Glutamate oxidation of 6C3HED lymphoma: effects of L-asparaginase on sensitive and resistant lines. Proc Natl Acad Sci USA. 1974;71:3993–3997.
  • Reitzer LJ, Wice BM, Kennell D. Evidence that glutamine, not sugar, is the major energy source for cultured HeLa cells. J Biol Chem. 1979;254:2669–2676.
  • Ardawi MS, Newsholme EA. Glutamine metabolism in lymphocytes of the rat. Biochem J. 1983;212:835–842.
  • Souba WW. Glutamine and cancer. Ann Surg. 1993;218(6):715–728.
  • Yang C, Ko B, Hensley CT, et al. Glutamine oxidation maintains the TCA cycle and cell survival during impaired mitochondrial pyruvate transport. Mol Cell. 2014;56:414–424.
  • Dennis B, Wikky T, VanNoorden CJ. Determination of phosphate-activated glutaminase activity and its kinetics in mouse tissues using metabolic mapping (quantitative enzyme histochemistry). J Histochem Cytochemistry Off J Histochem Soc. 2014;62(11):802–812.
  • Lane AN, Fan TW. Regulation of mammalian nucleotide metabolism and biosynthesis. Nucleic Acids Res. 2015;43:2466–2485.
  • Gaglio D, Soldati C, Vanoni M, et al. Glutamine deprivation induces abortive S-phase rescued by deoxyribonucleotides in k-ras transformed fibroblasts. Plos One. 2009;4:e4715.
  • Curi R, Newsholme P, Procopio J, et al. Glutamine, gene expression, and cell function. Front Biosci. 2007;12(1):344–357.
  • García-Maceira P, Mateo J. Silibinin inhibits hypoxia-inducible factor-1alpha and mTOR/p70S6K/4E-BP1 signalling pathway in human cervical and hepatoma cancer cells: implications for anticancer therapy. Oncogene. 2009;28(3):313–324.
  • Ploessl K, Wang L, Lieberman BP, et al. Comparative evaluation of 18F-labeled glutamic acid and glutamine as tumor metabolic imaging agents. J Nucl Med. 2012;53:1616–1624.
  • Lieberman BP, Ploessl K, Wang L, et al. PET imaging of glutaminolysis in tumors by 18F-(2S,4R)4-fluoroglutamine. J Nucl Med. 2011;52:1947–1955.
  • Kelloff GJ, Hoffman JM, Johnson B, et al. Progress and promise of FDG-PET imaging for cancer patient management and oncologic drug development. Clin Cancer Res. 2015;11:2785–2808.
  • Venneti S, Dunphy MP, Zhang H, et al. Glutamine-based PET imaging facilitates enhanced metabolic evaluation of gliomas in vivo.. Sci Transl Med. 2015;7:274ra17.
  • Choi C, Ganji S, Hulsey K, et al. A comparative study of short- and long-TE (1)H MRS at 3 T for in vivo detection of 2-hydroxyglutarate in brain tumors. NMR Biomed. 2013;26:1242–1250.
  • Marin-Valencia I, Yang C, Mashimo T, et al. Analysis of tumor metabolism reveals mitochondrial glucose oxidation in genetically diverse human glioblastomas in the mouse brain in vivo. Cell Metab. 2012;15:827–837.
  • Saverio T, Anaïs O, Ahmed SU, et al. Glutamine synthetase activity fuels nucleotide biosynthesis and supports growth of glutamine-restricted glioblastoma. Nat Cell Biol. 2015;17:1556–1568.
  • Lukey MJ, Wilson KF, Cerione RA. Therapeutic strategies impacting cancer cell glutamine metabolism. Future Med Chem. 2013;5:1685–1700.
  • Ap H, Jing J, Wooster RF, et al. Analysis of glutamine dependency in non-small cell lung cancer: GLS1 splice variant GAC is essential for cancer cell growth. Cancer Biol Ther. 2012;13:1185–1194.
  • Moreadith RW, Lehninger AL. The pathways of glutamate and glutamine oxidation by tumor cell mitochondria. Role of mitochondrial NAD(P)+ dependent malic enzyme. J Biol Chem. 1984;259:6215–6221.
  • Matés JM, Segura JA, Martín-Rufián M, et al. Glutaminase isoenzymes as key regulators in metabolic and oxidative stress against cancer. Curr Mol Med. 2013;13:514–534.
  • Curthoys NP, Watford M. Regulation of glutaminase activity and glutamine metabolism. Annu Rev Nutr. 1995;15:133–159.
  • Krebs HA. Metabolism of amino-acids: the synthesis of glutamine from glutamic acid and ammonia, and the enzymic hydrolysis of glutamine in animal tissues. Biochem. J. 1953;29:1951–1969.
  • Cassago A, Ferreira AP, Ferreira IM, et al. Mitochondrial localization and structure-based phosphate activation mechanism of glutaminase C with implications for cancer metabolism. Proc Natl Acad Sci USA. 2012;109:1092–1097.
  • Elgadi KM, Meguid RA, Qian M, et al. Cloning and analysis of unique human glutaminase isoforms generated by tissue-specific alternative splicing. Physiol Genom. 1999;1:51–62.
  • Shapiro RA, Farrell L, Srinivasan M, et al. Isolation, characterization, and in vitro expression of a cDNA that encodes the kidney isoenzyme of the mitochondrial glutaminase. J Biol Chem. 1991;266:18792–18796.
  • Ferreira AP, Cassago A, Gonçalves KA, et al. Active Glutaminase C Self-assembles into a Supratetrameric Oligomer That Can Be Disrupted by an Allosteric Inhibitor. J Biol Chem. 2013;288(39):28009–28020.
  • Turner A, McGivan JD. Glutaminase isoform expression in cell lines derived from human colorectal adenomas and carcinomas. Biochem J. 2003;370:403–408.
  • Szeliga M, Matyja E, Obara M, et al. Relative expression of mRNAS coding for glutaminase isoforms in CNS tissues and CNS tumors. Neurochem Res. 2008;33:808–813.
  • Szeliga M, Matyja E, Obara M, et al. Lack of expression of the liver-type glutaminase (LGA) mRNA in human malignant gliomas. Neuroscience Lett. 2005;374:171–173.
  • Altman BJ, Stine ZE, Cv D. From Krebs to clinic: glutamine metabolism to cancer therapy. Nat Rev Cancer. 2016 Dec;16(12):773.
  • Terunuma A, Putluri N, Mishra P, et al. MYC-driven accumulation of 2 hydroxyglutarate is associated with breast cancer prognosis. J Clin Invest. 2014;124:398–412.
  • Thai M, Thaker SK, Feng J, et al. MYC-induced reprogramming of glutamine catabolism supports optimal virus replication. Nat Commun. 2015;6:8873.
  • Sanchez EL, Carroll PA, Thalhofer AB, et al. Latent KSHV infected endothelial cells are glutamine addicted and require glutaminolysis for survival. PLoS Pathog. 2015;11:e1005052.
  • Chen Z, Wang Y, Warden C, et al. Cross-talk between ER and HER2 regulates cMYC-mediated glutamine metabolism in aromatase inhibitor resistant breast cancer cells. J Steroid Biochem Mol Biol. 2015;149:118–127.
  • Gaglio D, Metallo CM, Gameiro PA, et al. Oncogenic K-Ras decouples glucose and glutamine metabolism to support cancer cell growth. Mol Syst Biol. 2011;7:523.
  • Brunelli L, Caiola E, Marabese M, et al. Capturing the metabolomic diversity of KRAS mutants in non-small-cell lung cancer cells. Oncotarget. 2014;5:4722–4731.
  • Ping G, Irina T, Chang TC, et al. c-Myc suppression of miR-23a/b enhances mitochondrial glutaminase expression and glutamine metabolism. Nature. 2009;458:762–765.
  • Xiao D, Ren P, Su H, et al. Myc promotes glutaminolysis in human neuroblastoma through direct activation of glutaminase 2. Oncotarget. 2015;6(38):40655–40666.
  • Michael JL, Greene KS, Erickson JW, et al. The oncogenic transcription factor c-Jun regulates glutaminase expression and sensitizes cells to glutaminase-targeted therapy. Nat Commun. 2016;7:11321.
  • Zack TI, Schumacher SE, Carter SL, et al. Pan-cancer patterns of somatic copy number alteration. Nat Genet. 2103;45:1134–1140.
  • Sun RC, Denko NC. Hypoxic regulation of glutamine metabolism through HIF1 and SIAH2 supports lipid synthesis that is necessary for tumor growth. Cell Meta. 2014;19:285–292.
  • Gameiro PA, Yang J, Metelo AM, et al. In vivo HIF-mediated reductive carboxylation is regulated by citrate levels and sensitizes VHL-deficient cells to glutamine deprivation. Cell Metab. 2013;17:372–385.
  • Wise DR, Ward PS, Shay JE, et al. Hypoxia promotes isocitrate dehydrogenase-dependent carboxylation of α-ketoglutarate to citrate to support cell growth and viability. Proc Natl Acad Sci USA. 2011;108:19611–19616.
  • Wang JB, Erickson JW, Fuji R, et al. Targeting mitochondrial glutaminase activity inhibits oncogenic transformation. Cancer Cell. 2010;18:207–219.
  • Thangavelu K, Pan CQ, Karlberg T, et al. Structural basis for the allosteric inhibitory mMechanism of human kidney-type glutaminase (KGA) and its regulation by Raf-Mek-Erk signaling in cancer cell metabolism. Proc Natl Acad Sci USA. 2012;109:7705–7710.
  • Durán RV, Oppliger W, A M R, et al. Glutaminolysis activates Rag-mTORC1 signaling. Mol Cell. 2012;47:349–358.
  • Sancak Y, Peterson TR, Shaul YD, et al. The Rag GTPases bind raptor and mediate amino acid signaling to mTORC1. Science. 2008;320:1496–1501.
  • Thangavelu K, Chong QY, Low BC, et al. Structural basis for the active site inhibition mechanism of human kidney-type glutaminase (KGA). Sci Rep. 2014;4:3827.
  • Gross MI, Demo SD, Dennison JB, et al. Antitumor activity of the glutaminase inhibitor CB-839 in triple-negative breast cancer. Mol Cancer Ther. 2014;13(4):890–901.
  • Cheng L, Wu CR, Zhu LH, et al. Physapubescin, a natural withanolide as a kidney-type glutaminase (KGA) inhibitor. Bioorg Med Chem Lett. 2017;27(5):1243–1246.
  • Wu C, Zheng M, Gao S, et al. A natural inhibitor of kidney-type glutaminase: a withanolide from Physalis pubescens with potent anti-tumor activity. Oncotarget. 2017;8(69):113516–113530.
  • Newcomb RW, Newcomb M Selective inhibition of glutaminase by bis-thiadiazoles. US6451828B1 (2002).
  • Seltzer MJ, Bennett BD, Joshi AD, et al. Inhibition of glutaminase preferentially slows growth of glioma cells with mutant IDH1. Cancer Res. 2010;70(22):8981–8987.
  • Le A, Lane AN, Bose S, et al. Glucose-independent glutamine metabolism via TCA cycling for proliferation and survival in B-cells. Cell Metab. 2012;15(1):110–121.
  • DeLaBarre B, Gross S, Fang C, et al. Full-length human glutaminase in complex with an allosteric inhibitor. Biochemistry. 2011;50:10764–10770.
  • Liang Y, Bromley SD, Orford K Treatment of cancer with inhibitors of glutaminase. WO2018039442 (2018).
  • Huang Q, Katt WP, McDermott LA, et al. Crystal structure of glutaminase C in complex with inhibitor CB-839. PDB: 5HL1, 2016
  • Ramachandran S, Pan CQ, Zimmermann SC, et al. Structural basis for exploring the allosteric inhibition of human kidney type glutaminase. Oncotarget. 2016;7:57943–57954.
  • Matre P, Velez J, Jacamo R, et al. Inhibiting glutaminase in acute myeloid leukemia: metabolic dependency of selected AML subtypes. Oncotarget. 2016;7:79722–79735.
  • Parlati F, Demo SD, Gross MI, 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. Cancer Res. 2014;74:1416.
  • Parlati F, Gross M, Janes J, et al. Glutaminase inhibitor CB-839 synergizes with pomalidomide in preclinical multiple myeloma models. Blood. 2104;124:4720.
  • Parlati F, Works MG, Rodriguez M, et al. Treatment of lung cancer with inhibitors of glutaminase. SG11201708153X (2017)
  • Timothy FS, James JS, Jacqueline NW Crystal forms of glutaminase inhibitors. US20170226101 (2017)
  • Molineaux SM, Gross MI, Bromley SD, et al. Combination therapy with glutaminase inhibitors and immuo-oncology agent. WO2017062354 (2017)
  • Cerione R, Cerione K, Stalnecker C, et al. Inhibitors of kidney-type glutaminase, GLS-1. WO2016090350 (2016)
  • Robinson MM, McBryant SJ, Tsukamoto T, et al. Novel mechanism of inhibition of rat kidney-type glutaminase by bis-2-(5-phenylacetamido-1,2,4-thiadi -azol-2-yl) ethyl sulfide (BPTES). Biochem J. 2007;406(3):407–414.
  • Godfrey S, Kuhlenschmidt T, Curthoys P. Correlation between activation and dimer formation of rat renal phosphate-dependent glutaminase. J Biol Chem. 1997;252(6):1927–1931.
  • Kenny J, Bao Y, Hamm B, et al. Bacterial expression, purification, and characterization of rat kidney-type mitochondrial glutaminase. Protein Expr Purif. 2003;31(1):140–148.
  • Møller M, Niellsen SS, Ramachandran S, et al. Small angle X-ray scattering studies of mitochondrial glutaminase C reveal extended flexible regions, and link oligomeric state with enzyme activity. PLoS One. 2013;8(9):e74783.
  • Stalnecker CA, Ulrich SM, Li Y, et al. Mechanism by which a recently discovered allosteric inhibitor blocks glutamine metabolism in transformed cells. Proc Natl Acad Sci USA. 2015;112:394–399.
  • Gao M, Monian P, Quadri N,, et al. Glutaminolysis and Transferrin Regulate Ferroptosis. Mol Cell. 2015;59:298–308.
  • Son J, Lyssiotis CA, Ying H, et al. Glutamine supports pancreatic cancer growth through a KRAS-regulated metabolic pathway. Nature. 2013;496:101–105.
  • Simpson NE, Tryndyak VP, Pogribna M, et al. Modifying metabolically sensitive histone marks by inhibiting glutamine metabolism affects gene expression and alters cancer cell phenotype. Epigenetics. 2012;7:1413–1420.
  • Jacque N, Ronchetti AM, Larrue C, et al. Targeting glutaminolysis has antileukemic activity in acute myeloid leukemia and synergizes with BCL-2 inhibition. Blood. 2015;126:1346–1356.
  • Kahlert UD, Cheng M, Koch K, et al. Alterations in cellular metabolome after pharmacological inhibition of Notch in glioblastoma cells. Int J Cancer. 2012;138:1246–1255.
  • Yuan L, Sheng X, Clark LH, et al. Glutaminase inhibitor compound 968 inhibits cell proliferation and sensitizes paclitaxel in ovarian cancer. Am J Transl Res. 2016;8:4265–4277.
  • Mcdermott LA, Iyer PC, Cerione R, et al. Glutaminase inhibitors. WO2016054388 (2016)
  • Di L, Kerns EH, Hong Y, et al. Optimization of a higher throughput microsomal stability screening assay for profiling drug discovery candidates. J Biomol Screen. 2003;8(4):453–462.
  • Finlay M, Raymond V Bis-pyridazine compounds and their use in treating cancer. WO2017089587 (2017)
  • Finlay M, Raymond V, Nissink J, et al. 1,3,4-Thiadiazole compounds and their use in treating cancer. WO2017093299 (2017)
  • Ruan BF, Ruan J Selenium-containing inhibitors that bind to the allosteric site of glutaminase and/or inhibit glutamate dehydrogenase and their applications. WO2017084598 (2017)
  • Thomas AG, Rojas C, Tanega C, et al. Kinetic characterization of ebselen, chelerythrine and apomorphine as glutaminase inhibitors. Biochem Biophys Res Commun. 2013;438:243–248.
  • Emilia M, Jones P, Heffernan T, et al. GLS1 inhibitors for treating disease. US20170291895, 2017
  • Hanes J, Slusher BS, Le A, et al. Salt forms and polymorphs of (R)-1-(4-(6-(2-(4-(3,3-difluorocyclobutoxy)-6-methyloyridin-2-yl) acetamido) pyridazin −3-yl)-2-fluorobutyl)-N-methyl-1H-1,2,3-triazole-4-carboxamide. WO2017112831 (2016)
  • Agios Pharmaceuticals, Inc. Compounds and methods of use. AU2018202954 (2018)
  • Justin H, Barbara S, Anne L, et al. Glutaminase inhibitor discovery and nanoparticle-enhanced delivery for cancer therapy. W2016007647 (2016)
  • Elgogary A, Xu Q, Poore B, et al. Combination therapy with BPTES nanoparticles and metformin targets the metabolic heterogeneity of pancreatic cancer. Proc Natl Acad Sci USA. 2016;113(36):E5328–E5336.
  • Bausch M, Wetzler R, Muller C, et al. GlutaDON. WO2007128588 (2007)
  • McGregor WG, Roberts J. Glutaminase enhances therapeutic effectiveness of glutamine antimetabolites against human and murine solid tumors In Vivo. Am Assoc Cancer Res. 1989;30:p578.
  • Unger C, Harzmann R, Müller C, et al. Phase I dose escalating study of PEG-PGA and DON (GlutaDON): a new amino acid depleting anti cancer drug approach. J Clin Oncol. 2004;22:p3175.
  • Unger C, Harzmann R, Müller C, et al. Phase I dose escalating study of PEG-PGA and DON: a new amino acid depleting anti cancer drug approach. J Clin Oncol. 2005;23:p3130.
  • Boysen G, Griffin RJ Method of increasing radiation sensitivity of tumor cells. US20180104336 (2018)
  • Hou YL, Yuan LY Anti-tumor glutaminase inhibitor, tumor angiogenesis inhibitor drug compound and application there of. CN106890184 (2015)
  • Matés JM, Campos-Sandoval JA, Márquez J. Glutaminase isoenzymes in the metabolic therapy of cancer. Biochim Biophys Acta Rev Cancer. 2018;1870:158–164.
  • Merk A, Bartesaghi A, Banerjee S, et al. Breaking Cryo-EM Resolution Barriers to Facilitate Drug Discovery. Cell. 2016;165(7):1698–1707.
  • Harding JJ, Telli ML, Munster PN, et al. Safety and tolerability of increasing doses of CB-839, a first-in-class, orally administered small molecule inhibitor of glutaminase, in solid tumors. J Clin Oncol. 2015;33(15):2512.
  • Li J, Csibi A, Yang S, et al. Synthetic lethality of combined glutaminase and Hsp90 inhibition in mTORC1-driven tumor cells. Proc Natl Acad Sci USA. 2015;112:E21–E29.
  • Tanaka K, Sasayama T, Irino Y, et al. Compensatory glutamine metabolism promotes glioblastoma resistance to mTOR inhibitor treatment. J Clin Investig. 2015;125(4):1591.
  • J E H-A, T Q T, M A R, et al. Vemurafenib resistance reprograms melanoma cells towards glutamine dependence. J Transl Med. 2015;13(1):210.
  • Strohecker AM, Guo JY, Karsliuzunbas G, et al. Autophagy sustains mitochondrial glutamine metabolism and growth of BrafV600E-driven lung tumors. Autophagy. 2013;3(11):1272–1285.
  • Katt WP, Antonyak MA, Cerione RA. Simultaneously targeting tissue transglutaminase and kidney type glutaminase sensitizes cancer cells to acid toxicity and offers new opportunities for therapeutic intervention. Mol Pharm. 2015;12:46–55.
  • Xie C, Jin J, Bao X, et al. Inhibition of mitochondrial glutaminase activity reverses acquired erlotinib resistance in non-small cell lung cancer. Oncotarget. 2016;7(1):610–621.
  • Herranz D, Ambesiimpiombato A, Sudderth J, et al. Metabolic reprogramming induces resistance to anti-NOTCH1 therapies in acute lymphoblastic leukemia. Nature Medicine. 2015;21(10):1182–1189.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.