We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Targeting the latest hallmark of cancer: another attempt at ‘magic bullet’ drugs targeting cancers’ metabolic phenotype

    M Cuperlovic-Culf

    * Author for correspondence

    National Research Council of Canada, Institute for Information Technology, 100 des Aboiteaux Street, Moncton, NB, E1A 7R1, Canada.

    ,
    AS Culf

    Atlantic Cancer Research Institute, 35 Providence Street, Moncton, NB, E1C 8X3, Canada

    ,
    M Touaibia

    Département de Chimie et Biochimie, Université de Moncton, 18, Avenue Antonine Maillet Moncton, NB, E1A 3E9, Canada

    &
    N Lefort

    Atlantic Cancer Research Institute, 35 Providence Street, Moncton, NB, E1C 8X3, Canada

    Published Online:https://doi.org/10.2217/fon.12.121

    The metabolism of tumors is remarkably different from the metabolism of corresponding normal cells and tissues. Metabolic alterations are initiated by oncogenes and are required for malignant transformation, allowing cancer cells to resist some cell death signals while producing energy and fulfilling their biosynthetic needs with limiting resources. The distinct metabolic phenotype of cancers provides an interesting avenue for treatment, potentially with minimal side effects. As many cancers show similar metabolic characteristics, drugs targeting the cancer metabolic phenotype are, perhaps optimistically, expected to be ‘magic bullet’ treatments. Over the last few years there have been a number of potential drugs developed to specifically target cancer metabolism. Several of these drugs are currently in clinical and preclinical trials. This review outlines examples of drugs developed for different targets of significance to cancer metabolism, with a focus on small molecule leads, chemical biology and clinical results for these drugs.

    Papers of special note have been highlighted as: ▪ of interest ▪▪ of considerable interest

    References

    • Cairns R, Harris IS, Mak TW. Regulation of cancer cell metabolism. Nat. Rev. Cancer11,85–95 (2011).▪▪ Detailed review of Warburg effect causes and consequences.
    • Kroemer G, Pouyssegur J. Tumor cell metabolism: cancer’s Achilles’ heel. Cancer Cell13,472–482 (2008).
    • Allen M, Jones JL. Jekyll and Hyde: the role of the microenvironment on the progression of cancer. J. Pathol.223,162–176 (2011).
    • Tennant D, Duran RV, Gottlieb E. Targeting metabolic transformation for cancer therapy. Nat. Rev. Cancer10,267–277 (2010).
    • Purcell WT, Ettinger DS. Novel antifolate drugs. Curr. Oncol. Rep.5,114–125 (2003).
    • Ramos-Montoya A, Lee WN, Bassilian S et al. Pentose phosphate cycle oxidative and nonoxidative balance: a new vulnerable target for overcoming drug resistance in cancer. Int. J. Cancer119,2733–2741 (2006).
    • Vizan P, Alcarraz-Vizan A, Diaz-Moralli S, Solovjeva O, Frederiks WM, Cascante M. Modulation of pentose phosphate pathway during cell cycle progression in human colon adenocarcinoma cell line HT29. Int. J. Cancer124,2789–2796 (2009).
    • Porporato PE, Dhup S, Dadhich RK, Copetti T, Sonveaux P. Anticancer targets in the glycolytic metabolism of tumors: a comprehensive review. Front. Pharm.2,1–18 (2011).
    • Cao X, Fang L, Gibbs S et al. Glucose uptake inhibitor sensitizes cancer cells to daunorubicin and overcomes drug resistance in hypoxia. Cancer Chemo Pharm.59,495–505 (2007).
    • 10  Flaig TW, Glodé M, Gustafson D et al. A study of high-dose oral silybin-phytosome followed by prostatectomy in patients with localized prostate cancer. Prostate70(8),848–855 (2010).
    • 11  Zhang W, Liu Y, Chen X, Bergmeier SC. Novel inhibitors of basal glucose transport as potential anticancer agents. Bioorg. Med. Chem. Lett.20,2191–2194 (2010).
    • 12  Chan DA, Sutphin PD, Nguyen P et al. Targeting GLUT1 and the warburg effect in renal cell carcinoma by chemical synthetic lethality. Sci. Transl. Med.3,94ra70 (2011).
    • 13  Pelicano H, Martin DS, Xu RH, Huang P. Glycolysis inhibition for anticancer treatment. Oncogene25,4633–4646 (2006).
    • 14  Chen W, Guéron M. The inhibition of bovine heart hexokinase by 2-deoxy-D-glucose-6-phosphate: characterization by 31P NMR and metabolic implications. Biochimie74,867–873 (1992).
    • 15  Landau BR, Laszlo J, Stengle J, Burk D. Certain metabolic and pharmacologic effects in cancer patients given infusions of 2-deoxy-D-glucose. J. Natl Cancer Inst.21,485–494 (1958).
    • 16  Atsumi T, Chesney J, Metz C et al. High expression of inducible 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (iPFK-2; PFKFB3) in human cancers. Cancer Res.62,5881–5887 (2002).
    • 17  Clem B, Telang S, Clem A et al. Small-molecule inhibition of 6-phosphofructo-2-kinase activity suppresses glycolytic flux and tumor growth. Mol. Cancer Ther.7,110–120 (2008).
    • 18  Mazurek S, Boschek CB, Hugo F, Eigenbrodt E. Pyruvate kinase type M2 and its role in tumor growth and spreading. Semin. Cancer Biol.15,300–308 (2005).
    • 19  Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science324,1029–1033 (2009).
    • 20  Vander Heiden MG, Christofk HR, Schuman E et al. Identification of small molecule inhibitors of pyruvate kinase M2. Biochem. Pharmacol.79,1118–1124 (2010).
    • 21  Kim JW, Tchernyshyov I, Semenza GL, Dang CV. HIF-1-mediated expression of pyruvate dehydrogenase kinase: a metabolic switch required for cellular adaptation to hypoxia. Cell Metab.3,177–185 (2006).▪▪ Describes the effect of HIF-1 on establishment of tumor metabolic phenotype.
    • 22  Papandreou I, Cairns RA, Fontana L, Lim AL, Denko NC. HIF-1 mediates adaptation to hypoxia by actively down regulating mitochondrial oxygen consumption. Cell Metab.3,187–197 (2006).
    • 23  Whitehouse S, Cooper RH, Randle PJ. Mechanism of activation of pyruvate dehydrogenase by dichloroacetate and other halogenated carboxylic acids. Biochem. J.141,761–774 (1974).
    • 24  Bonnet S, Archer SL, Allalunis-Turner J et al. A mitochondria-K+ channel axis is suppressed in cancer and its normalization promotes apoptosis and inhibits cancer growth. Cancer Cell11,37–51 (2007).
    • 25  Michelakis ED, Sutendra G, Dromparis P et al. Metabolic modulation of glioblastoma with dichloroacetate. Sci. Trans. Med.2,31ra34 (2010).
    • 26  Aicher TD, Damon RE, Koletar J et al. Triterpene and diterpene inhibitors of pyruvate dehydrogenase kinase (PDK). Bioorg. Med. Chem. Lett.9,2223–2228 (1999).
    • 27  Aicher TD, Anderson RC, Gao J et al. Secondary amides of (R)-3,3,3-trifluoro-2-hydroxy-2-methylpropionic acid as inhibitors of pyruvate dehydrogenase kinase. J. Med. Chem.43,236–249 (2000).
    • 28  Bebernitz GR, Aicher TD, Stanton JL et al. Anilides of (R)-trifluoro-2-hydroxy-2-methylpropionic acid as inhibitors of pyruvate dehydrogenase kinase. J. Med. Chem.43,2248–2257 (2000).
    • 29  Sonveau P, Vegran F, Schoroeder T et al. Targeting lactate-fueled respiration selectively kills hypoxic tumor cells in mice. J. Clin. Invest.118,3930–3942 (2008).▪▪ First publication showing a controversial hypothesis regarding function of lactic acid production in tumor cells.
    • 30  Le A, Cooper CR, Gouw AM et al. Inhibition of lactate dehydrogenase A induces oxidative stress and inhibits tumor progression. Proc. Natl Acad. Sci. USA107,2037–2042 (2010).
    • 31  Granchi C, Bertini S, Macchia M, Minutolo F. Inhibitors of lactate dehydrogenase isoforms and their therapeutic potentials. Cur. Med. Chem.17,672–697 (2010).
    • 32  Granchi C, Roy S, De Simone A et al. N-Hydroxyindole-based inhibitors of lactate dehydrogenase against cancer cell proliferation. Eur. J. Med. Chem.46,5398–5407 (2011).
    • 33  Kroemer G. Mitochondria in cancer. Oncogene25,4630–4632 (2011).▪▪ Detailed review of the functions and significance of mitochondria and mitochondria-based processes in cancer development and progression.
    • 34  Warburg O. On the origin of cancer cells. Science123,309–314 (1956).
    • 35  Vazquez A, Oltvai ZN. Molecular crowding defines a common origin for the Warburg effect in proliferating cells and the lactate threshold in muscle physiology. PLoS ONE6,e19538 (2011).
    • 36  Bhalla K, Hwang BJ, Dewi RE et al. PGC1α promotes tumor growth by inducing gene expression programs supporting lipogenesis. Cancer Res.71,6888–6898 (2011).
    • 37  D’Errico I, Salvatore L, Murzilli S et al. Peroxisome proliferator-activated receptor-γ coactivator 1-α (PGC1α) is a metabolic regulator of intestinal epithelial cell fate. Proc. Natl Acad. Sci. USA108,6603–6608 (2011).
    • 38  Wang X, Moraes CT. Increases in mitochondrial biogenesis impair carcinogenesis at multiple levels. Mol. Oncol.5,399–409 (2011).
    • 39  Guan KL, Xiong Y. Regulation of intermediary metabolism by protein acetylation. Trends Biochem. Sci.36,108–116 (2011).
    • 40  Amoêdo ND, Rodrigues MF, Pezzuto P et al. Energy metabolism in H460 lung cancer cells: effects of histone deacetylase inhibitors. PLoS ONE6,e22264 (2011).
    • 41  Hirschey MD, Shimazu T, Jing E et al. SIRT3 deficiency and mitochondrial protein hyperacetylation accelerate the development of the metabolic syndrome. Mol. Cell44,177–190 (2011).
    • 42  Kourelis TV, Siegel RD. Metformin and cancer: new applications for an old drug. Med. Oncol.29(2),1314–1327 (2011).▪ Detailed consideration of the potential for use of metformin, a widely available hypoglycemic agent, for anticancer treatment.
    • 43  Biasutto L, Dong LF, Zoratti M, Neuzil J. Mitochondrially targeted anti-cancer agents. Mitochondrion10,670–681 (2010).
    • 44  Dong LF, Freeman R, Liu J et al. Suppression of tumor growth in vivo by the mitocan α-tocopheryl succinate requires respiratory complex II. Clin. Cancer Res.15,1593–1600 (2009).
    • 45  Raj L, Ide T, Gurkar AU et al. Selective killing of cancer cells by a small molecule targeting the stress response to ROS. Nature475,231–234 (2011).▪ Interesting example of the effect of a drug leading to an increase in reactive oxygen species level and the initiation of apoptotic cell death in cancer cells.
    • 46  Stumvoll M, Nurjhan N, Perriello G, Dailey G, Gerich JE. Metabolic effects of metfomin in non-insulin-dependent diabetes mellitus. New Eng. J. Med.333,550–554 (1995).
    • 47  Hundal HS, Ramlal T, Reyes R, Leiter LA, Klip A. Cellular mechanism of metformin action involves glucose transporter translocation from an intracellular pool to the plasma membrane in L6 muscle cells. Endocrinology131,1165–1173 (1992).
    • 48  Owen M, Doran E, Halestrap AP. Evidence that metformin exerts its anti-diabetic effects through inhibition of complex I of the mitochondrial respiratory chain. Biochem. J.348,607–614 (2000).
    • 49  Shaw RJ, Kosmatka M, Bardessey N et al. The tumor supressor LKB1 kinase directly activates AMP-activated kinase and regulates apoptosis in response to energy stress. Proc. Natl Acad. Sci. USA101,3329–3335 (2004).
    • 50  Pierotti MA, Berrino F, Gariboldi M et al. Targeting metabolism for cancer treatment and prevention: metformin, an old drug with multi-faceted effects. Oncogene doi:10.1038/onc.2012.181 (2012) (Epub ahead of print).
    • 51  Evans JM, Donnelly LA, Emslie-Smith AM, Alessi DR, Morris AD. Metformin and reduced risk of cancer in diabetic patients. Br. Med. J.330,1304–1305 (2005).
    • 52  Bowker SL, Majumdar SR, Veugelers P, Johnson JA. Increased cancer-related mortality for patients with type 2 diabetes who use sulfonylureas or insulin. Diabetes Care29,254–258 (2006).
    • 53  Blandino G, Valerio M, Cioce M et al. Metformin elicits anticancer effects through the sequential modulation of DICER and c-MYC. Nat. Commun.3,865 (2012).
    • 54  Wu B, Li S, Sheng L et al. Metformin inhibits the development and metastasis of ovarian cancer. Oncol. Reprod.28,903–908 (2012).
    • 55  Martin M, Marais R. Metformin: a diabetes drug for cancer, or a cancer drug for diabetics? J. Clin. Oncol.30(21),2698–2700 (2012).
    • 56  Zaugg K, Yao Y, Reilly PT et al. Carnitine palmitoyltransferase 1C promotes cell survival and tumor growth under conditions of metabolic stress. Genes Dev.25,1041–1051 (2011).
    • 57  Zhou W, Tu Y, Simpson PJ, Kuhajda FP. Malonyl-CoA decarboxylase inhibition is selectively cytotoxic to human breast cancer cells. Oncogene28,2979–2987 (2009).
    • 58  Nieman KM, Kenny HA, Penicka CV et al. Adipocytes promote ovarian cancer metastasis and provide energy for rapid tumor growth. Nat. Med.17,1498–1503 (2011).
    • 59  Das SK, Eder S, Schauer S et al. Adipose triglyceride lipase contributes to cancer-associated cachexia. Science333(6039),233–238 (2011).
    • 60  Swinnen JV, Van Veldhoven PP, Timmermans L et al. Fatty acid synthase drives the synthesis of phospholipids partitioning into detergent-resistant membrane microdomains. Biochem. Biophys. Res. Commun.302,898–903 (2003).
    • 61  Menendez JA, Lupu R. Fatty acid synthase and the lipogenic phenotype in cancer pathogenesis. Nat. Rev. Cancer7,763–777 (2007).
    • 62  Mashima T, Seimiya H, Tsuruo T. De novo fatty-acid synthesis and related pathways as molecular targets for cancer therapy. Br. J. Cancer100,1369–1372 (2009).
    • 63  Migita T, Narita T, Nomura K et al. ATP citrate lyase: activation and therapeutic implications in non-small cell lung cancer. Cancer Res.68,8547–8554 (2008).
    • 64  Hatzivassiliou G, Zhao F, Bauer DE et al. ATP citrate lyase inhibition can suppress tumor cell growth. Cancer Cell8,311–321 (2005).
    • 65  Beckner ME, Fellows-Mayle W, Zhang Z et al. Identification of ATP citrate lyase as a positive regulator of glycolytic function in glioblastomas. Int. J. Cancer126,2282–2295 (2010).
    • 66  Pearce NJ, Yates JW, Berkhout TA et al. The role of ATP citrate-lyase in the metabolic regulation of plasma lipids. Hypolipidaemic effects of SB-204990, a lactone prodrug of the potent ATP citrate-lyase inhibitor SB-201076. Biochem. J.334,113–119 (1998).
    • 67  López-Lázaro M. The Warburg effect: why and how do cancer cells activate glycolysis in the presence of oxygen? Anticancer Agents Med. Chem.8,305–312 (2008).
    • 68  Pappenberger G, Benz J, Gsell B et al. Structure of the human fatty acid synthase KS-MAT didomain as a framework for inhibitor design. J. Mol. Biol.397,508–519 (2010).
    • 69  De Schrijver E, Brusselmans K, Heyns W, Verhoeven G, Swinnen JV. RNA interference-mediated silencing of the fatty acid synthase gene attenuates growth and induces morphological changes and apoptosis of LNCaP prostate cancer cells. Cancer Res.63,3799–3804 (2003).
    • 70  Kridel SJ, Lowther WT, Pemble CW 4th. Fatty acid synthase inhibitors: new directions for oncology. Expert Opin Invest. Drugs16,1817–1829 (2007).
    • 71  Price AC, Choi KH, Heath RJ, Li Z, White SW, Rock CO. Inhibition of β-ketoacyl-acyl carrier protein synthases by thiolactomycin and cerulenin. Structure and mechanism. J. Biol. Chem.276,6551–6559 (2001).
    • 72  Moche M, Schneider G, Edwards P, Dehesh K, Lindqvist Y. Structure of the complex between the antibiotic cerulenin and its target, β-ketoacyl-acyl carrier protein synthase. J. Biol. Chem.274,6031–6034 (1999).
    • 73  Wang X, Tian W. Green tea epigallocatechin gallate: a natural inhibitor of fatty acid synthase. Biochem. Biophys. Res. Commun.288,1200–1206 (2001).
    • 74  Menendez JA, Colomer R, Lupu R. Inhibition of fatty acide synthase-dependent neoplastic lipogenesis as the mechanism of γ-linolenic acid-induced toxicity to tumor cells: an extension to Nwankwo’s hypothesis. Med. Hypothesis64,337–341 (2005).
    • 75  Vázquez MJ, Leavens W, Liu R et al. Discovery of GSK837149A, an inhibitor of human fatty acid synthase targeting the β-ketoacyl reductase reaction. FEBS J.275,1556–1567 (2008).
    • 76  Zhan Y, Ginanni N, Tota MR et al. Control of cell growth and survival by enzymes of the fatty acid synthesis pathway in HCT-116 colon cancer cells. Clin. Cancer Res.14,5735–5742 (2008).
    • 77  Brusselmans K, De Schrijver E, Verhoeven G, Swinnen JV. RNA interference-mediated silencing of the acetyl coA carboxylase-α gene induces growth inhibition and apoptosis of prostate cancer cells. Cancer Res.65,6719–6725 (2005).
    • 78  Waldrop GL, Stephens JM. Targeting acetyl coA carboxylase for anti-obesity therapy. Curr. Med. Chem. Immunol. Endocr. Metabol. Agents3,229–234 (2003).
    • 79  Thupari JN, Pinn ML, Kuhajda FP. Fatty acid synthase inhibition in human breast cancer cells leads to malonyl coA induced inhibition of fatty acid oxidation and cytotoxicity. Biochem. Biophys. Res. Commun.285,217–223 (2001).
    • 80  Wang C, Xu C, Sun M, Luo D, Liao DF, Cao D. Acetyl coA carboxylase-α inhibitor TOFA induces human cancer cell apoptosis. Biochem. Biophys. Res. Commun.385,302–306 (2009).
    • 81  Sugimoto Y, Naniwa Y, Nakamura T et al. A novel acetyl-CoA carboxylase inhibitor reduces de novo fatty acid synthesis in HepG2 cells and rat primary hepatocytes. Archiv. Biochem. Biophys.468,44–48 (2007).
    • 82  Beckers A, Organe S, Timmermans L et al. Chemical inhibition of acetyl coA carboxylase induces growth arrest and cytotoxicity selectively in cancer cells. Cancer Res.67,8180–8187 (2007).
    • 83  Harwood HJ Jr, Petras SF, Shelly LD et al. Isozyme-nonselective N-substituted bipiperidylcarboxamide acetyl-CoA carboxylase inhibitors reduce tissue malonyl-CoA concentrations, inhibit fatty acid synthesis, and increase fatty acid oxidation in cultured cells and in experimental animals. J. Biol. Chem.278,37099–37111 (2003).
    • 84  Zhang H, Tweel B, Li J, Tong L. Crystal Structure of the carboxyltransferase domain of acetyl-coenzyme a carboxylase in complex with CP-640186. Structure12,1683–1692 (2004).
    • 85  Marjanovic J, Chalupska D, Patenode C et al. Recombinant yeast screen for new inhibitors of human acetyl-CoA carboxylase 2 identifies potential drugs to treat obesity. Proc. Natl Acad. Sci. USA107,9093–9098 (2010).
    • 86  Gu YG, Weitzberg M, Clark RF et al. Synthesis and structure – activity relationships of N-{3-[2-(4-Alkoxyphenoxy)thiazol-5-yl]-1- methylprop-2-ynyl}carboxy derivatives as selective acetyl-CoA carboxylase 2 inhibitors. J. Med. Chem.49,3770–3773 (2006).
    • 87  Vaklavas C, Chatzizisis YS, Tsimberidou AM. Common cardiovascular medications in cancer therapeutics. Pharmcol. Ther.130,177–190 (2011).
    • 88  Boudreau DM, Yu O, Johnson J. Statin use and cancer risk: a comprehensive review. Expert Opin Drug Saf.9,603–621 (2010).
    • 89  Mondul AM, Han M, Humphreys EB, Meinhold CL, Walsh PC, Platz EA. Association of statin use with pathological tumor characteristics and prostate cancer recurrence after surgery. J. Urol.185,1268–1273 (2011).
    • 90  Khurana V, Bejjanki HR, Caldito G, Owens MW. Statins reduce the risk of lung cancer in humans: a large case-control study of US veterans. Chest131,1282–1288 (2007).
    • 91  Dale KM, Coleman CI, Henyan NN, Kluger J, White CM. Statins and cancer risk. JAMA295,74–80 (2006).
    • 92  Jacobs EJ, Newton CC, Thun MJ, Gapstur SM. Long-term use of cholesterol-lowering drugs and cancer incidence in a large United States cohort. Cancer Res.71,1763–1771 (2011).▪ Presentation of the results of statin use in an extremely large, longitudinal cohort of patients.
    • 93  Clendening JW, Pandyra A, Boutros PC et al. Dysregulation of the mevalonate pathway promotes transformation. Proc. Natl Acad. Sci. USA107,15051–15056 (2010).
    • 94  Halestrap AP. The monocarboxylate transporter family-Structure and functional characterization. IUBMB Life64,1–9 (2012).
    • 95  Zheng XT, Yang HB, Li CM. Optical detection of single cell lactate release for cancer metabolic analysis. Anal. Chem.82,5082–5087 (2010).
    • 96  Semenza GL. Tumor metabolism: cancer cells give and take lactate. J. Clin. Invest.118,3835–3837 (2008).
    • 97  Gillies RJ, Robey I, Gatenby RA. Causes and consequences of increased glucose metabolism of cancers. J. Nucl. Med.49,S24–S42 (2008).
    • 98  Robey IF, Baggett BK, Kirkpatrick D et al. Bicarbonate increases tumor pH and inhibits spontaneous metastases. Cancer Res.69,2260–2268 (2009).
    • 99  Végran F, Boidot R, Michiels C, Sonveaux P, Feron O. Lactate influx through the endothelial cell monocarboxylate transporter MCT1 supports an NF-κB/IL-8 pathway that drives tumor angiogenesis. Cancer Res.71,2550–2560 (2011).
    • 100  Shim CK, Cheon EP, Kang KW, Seo KS, Han HK. Inhibition effect of flavonoids on monocarboxylate transporter 1 (MCT1) in Caco-2 cells. J. Pharm. Pharmacol.59,1515–1519 (2007).
    • 101  Wang X, Levi AJ, Halestrap AP. Substrate and inhibitor specificities of the monocarboxylate transporters of single rat heart cells. Am. J. Physiol. Heart Circ. Physiol.270,H476–H484 (1996).
    • 102  Melena J, Safa R, Graham M, Casson RJ, Osborne NN. The monocarboxylate transport inhibitor, α-cyano-4-hydroxycinnamate, has no effect on retinal ischemia. Brain Res.989,128–134 (2003).
    • 103  Murray CM, Hutchinson R, Bantick JR et al. Monocarboxylate transporter MCT1 is a target for immunosuppression. Nat. Chem. Biol.1,371–376 (2005).
    • 104  Bueno V, Binet I, Steger U et al. The specific monocarboxylate transporter (MCT1) inhibitor, AR C117977, a novel immunosuppressant, prolongs allograft survival in the mouse. Transplantation84(9),1204–1207 (2007).
    • 105  Ovens MJ, Davies AJ, Wilson MC, Murray CM, Halestrap AP. AR-C155858 is a potent inhibitor of monocarboxylate transporters MCT1 and MCT2 that binds to an intracellular site involving transmembrane helices 7–10. Biochem. J.425(3),523–530 (2010).
    • 106  Izumi H, Takahashi M, Uramoto H et al. Monocarboxylate transporters 1 and 4 are involved in the invasion activity of human lung cancer cells. Cancer Sci.102,1007–1013 (2011).
    • 107  Supuran CT. Carbonic anhydrase inhibitors. Bioorg. Med. Chem. Lett.20,3467–3474 (2010).
    • 108  Poulsen S. Carbonic anhydrase inhibition as a cancer therapy: a review of patent literature, 2007–2009. Expert Opin Ther. Pat.20(6),795–806 (2010).
    • 109  Alterio V, Hilvo M, Di Fiore A et al. Crystal structure of the catalytic domain of the tumor-associated human carbonic anhydrase IX. Proc. Natl Acad. Sci. USA106,16233–16238 (2009).
    • 110  Buller F, Steiner M, Frey K et al. Selection of carbonic anhydrase ix inhibitors from one million DNA-encoded compounds. ACS Chem. Biol.6,336–344 (2011).▪▪ Example of a very interesting novel approach for barcoding and testing a large number of chemical compounds for a particular application (in this case inhibition of carbonic anhydrase).
    • 111  Pacchiano F, Carta F, McDonald PC et al. Ureido-substituted benzenesulfonamides potently inhibit carbonic anhydrase IX and show antimetastatic activity in a model of breast cancer metastasis. J. Med. Chem.54,1896–1902 (2011).
    • 112  Dittrich C, Zandvliet AS, Gneist M, Huitema AD, King AA, Wanders J. A Phase I and pharmacokinetic study of indisulam in combination with carboplatin. Br. J. Cancer96,559–566 (2007).
    • 113  Talbot DC, von Pawel J, Cattell E et al. A randomized Phase II pharmacokinetic and pharmacodynamic study of indisulam as second-line therapy in patients with advanced non-small cell lung cancer. Clin. Cancer Res.13,1816–1822 (2007).
    • 114  Samid D, Yeh A, Prasanna P. Induction of erythroid differentiation and fetal hemoglobin production in human leukemic cells treated with phenylacetate. Blood80,1576–1481 (1992).
    • 115  Gao P, Tchernyshyov I, Chang TC et al. c-Myc suppression of miR-23a/b enhances mitochondrial glutaminase expression and glutamine metabolism. Nature458,762–765 (2009).
    • 116  Seltzer MJ, Bennett BD, Joshi AD et al. Inhibition of glutaminase preferentially slows growth of glioma cells with mutant IDH1. Cancer Res.70,8981–8987 (2010).
    • 117  Wang JB, Erickson JW, Fuji R et al. Targeting mitochondrial glutaminase activity inhibits oncogenic transformation. Cancer Cell18,207–219 (2010).
    • 118  Rosenfeld H, Roberts J. Enhancement of antitumour activity of glutamine antagonists 6-diazo-5-oxo-l-norleucine and acivicin in cell culture by glutamineasparginase. Cancer Res.41,1324–1328 (1981).
    • 119  Masetti R, Pession A. First-line treatment of acute lymphoblastic leukemia with peg asparaginase. Biologics3,359–368 (2009).
    • 120  Haskell CM, Canellos GP. L-asparaginase resistance in human leukemia–asparagine synthetase. Biochem. Pharmacol.18,2578–2580 (1969).
    • 121  Haskell CM, Canellos GP, Leventhal BG et al. L-asparaginase: therapeutic and toxic effects in patients with neoplastic disease. N. Engl. J. Med.281,1028–1034 (1969).
    • 122  Wheatley DN. Controlling cancer by restricting arginine availability – arginine-catabolizing enzymes as anticancer agents. Anticancer Drugs15,825–833 (2004).
    • 123  Ni Y, Schwaneberg U, Sun ZH. Arginine deiminase, a potential anti-tumor drug. Cancer Lett.261,1–11 (2008).
    • 124  Kanehisa M, Goto S, Kawashima S, Okuno Y, Hattori M. The KEGG resource for deciphering the genome. Nucleic Acids Res.32(Database issue),D277–D280 (2004).
    • 201  THE JOHN HOPKINS UNIVERSITY: WO2008076964 (2008).
    • 202  THE JOHN HOPKINS UNIVERSITY: WO2006042062 (2006).
    • 203  Lakatos GC: US4620972 (1986).
    • 204  UNIVERSITÀ DI PISA: WO2011054525 (2011).
    • 301  SciFinder®. www.cas.org/products/scifinder
    • 302  Cancer Research UK Press Release (2010). www.cancerresearchuk.org/cancer-info/news/archive/pressrelease/2010-09-13-%20cancer-drug-astrazeneca
    • 303  NIH. ClinicalTrials.gov. www.clinicaltrials.gov