1,480
Views
126
CrossRef citations to date
0
Altmetric
Review

Glycolytic enzyme inhibitors in cancer treatment

, , , &
Pages 1533-1545 | Published online: 22 Sep 2008

Bibliography

  • Chu E, De Vita VT. Principles of cancer management: chemotherapy. In: De Vita VT, Hellman S, Rosenberg SA, editors, Cancer – principles & practice of oncology. 6th edition. Philadelphia USA. Lippincot Williams and Wilkins; 2001. p. 224-35
  • Verweij J, Nooter K, Stoter G. Principles of chemotherapy. In: Souhami RL, Tannock I, Hohenberger P, Horiot JC, editors, Oxford textbook of oncology. 2nd edition. Oxford University Press; 2002. p. 404-12
  • Kitano H. Cancer as a robust system: implications for anticancer therapy. Nat Rev Cancer 2004;4:227-35
  • Allen TM. Ligand-targeted therapeutics in anticancer therapy. Nat Rev Cancer 2002;2:750-63
  • Gatenby RA, Gillies RJ. Why do cancers have high aerobic glycolysis? Nat Rev Cancer 2004;4:891-9
  • Warburg O. On the origin of cancer cells. Science 1956;123:309-14
  • Hammerman PS, Fox CJ, Thompson CB. Beginnings of a signal-transduction pathway for bioenergetic control of cell survival. Trends Biochem Sci 2004;29:586-92
  • Moreno-Sanchez R, Rodriguez-Enriquez S, Marin-Hernandez A, Saavedra E. Energy metabolism in tumor cells. FEBS J 2007;274:1393-418
  • Kim JW, Dang CV. Cancer's molecular sweet tooth and the Warburg effect. Cancer Res 2007;66:8927-30
  • Garber K. Energy deregulation: licensing tumor to grow. Science 2006;312:1158-9
  • Kim JW, Dang CV. Multifaceted roles of glycolytic enzymes. Trends Biochem Sci 2005;30:142-50
  • Kondoh H, Lleonart ME, Gil J, et al. Glycolytic enzymes can modulate cellular life span. Cancer Res 2005;65:177-85
  • Gottschalk S, Anderson N, Hainz C, et al. Imatinib (STI571) – mediated changes in glucose metabolism in human leukemia BCR-ABL-positive cells. Clin Cancer Res 2004;10:6661-8
  • Costello LC, Franklin RB. ‘Why do tumour cells glycolyse?’: from glycolysis through citrate to lipogenesis. Mol Cell Biochem 2005;280:1-8
  • Shaw RJ. Glucose metabolism and cancer. Curr Opin Cell Biol 2006;18:598-608
  • Kroemer G, Pouyssegur J. Tumor cell metabolism: cancer's Achilles' heel. Cancer Cell 2008;13:472-82
  • Deberardinis RJ, Sayed N, Ditsworth D, Thompson CB. Brick by brick: metabolism and tumor cell growth. Curr Opin Genet Dev 2008;18:54-61
  • Deberardinis RJ, Lum JJ, Hatzivassiliou G, Thompson CB. The biology of cancer: metabolic reprogramming fuels cell growth and proliferation. Cell Metab 2008;7:11-20
  • Naumov GN, Bender E, Zurakowski D, et al. A model of human tumor dormancy: an angiogenic switch from the nonangiogenic phenotype. J Natl Cancer Inst 2006;98:316-25
  • Yasuda M, Matsubara J, Yamasaki H, et al. Death-resistant and nonresistant malignant human cell lines under anoxia in vitro. Int J Clin Oncol 2007;12:455-62
  • Brahimi-Horn MC, Chiche J, Pouysségur J. Hypoxia and cancer. J Mol Med 2007;85:1301-7
  • Bristow RG, Hill RP. Hypoxia and metabolism. Hypoxia, DNA repair and genetic instability. Nat Rev Cancer 2008;8:180-92
  • Dewhirst MW, Cao Y, Moeller B. Cycling hypoxia and free radicals regulate angiogenesis and radiotherapy response. Nat Rev Cancer 2008;8:425-37
  • Pelicano H, Martin DS, Xu RH, Huang P. Glycolysis inhibition for anticancer treatment. Oncogene 2006;25:4633-46
  • Chen Z, Lu W, Garcia-Prieto C, Huang P. The Warburg effect and its cancer therapeutic implications. J Bioenerg Biomembr 2007;39:267-74
  • Kimura Y, Morita SY, Matsuo M, Ueda K. Mechanism of multi-drug recognition by MDR1/ABCB1. Cancer Sci 2007;98:1303-10
  • Leslie EM, Deeley RG, Cole SP. Multi-drug resistance proteins: role of P-glycoprotein, MRP1, MRP2, and BCRP (ABCG2) in tissue defense. Toxicol Appl Pharmacol 2005;204:216-37
  • Stubbs E, Kraas J, Morton KA, Clark PB. Brain abnormalities detected on whole-body 18F-FDG PET in cancer patients: spectrum of findings. AJR Am J Roentgenol 2007;188:866-73
  • Wienhard K. Measurement of glucose consumption using [(18)F]fluorodeoxyglucose. Methods 2002;27:218-25
  • Van der Hiel B, Pauwels EK, Stokkel MP. Positron emission tomography with 2-[18F]-fluoro-2-deoxy-D-glucose in oncology. Part IIIa. Therapy response monitoring in breast cancer, lymphoma and gliomas. J Cancer Res Clin Oncol 2001;127:269-77
  • Harris RA. Carbohydrate metabolism I: major metabolic pathways and their control. In: David L, Devlin TM, editiors, Textbook of biochemistry with clinical correlations. 6th edition. John Wiley & Sons, Inc. Hoboken, NJ; 2006. p. 581-635
  • Mathupala SP, Ko YH, Pedersen PL. Hexokinase II: cancer's double-edged sword acting as both facilitator and gatekeeper of malignancy when bound to mitochondria. Oncogene 2006;25:4777-86
  • Robey RB, Hay N. Mitochondrial hexokinases, novel mediators of the antiapoptotic effects of growth factors and Akt. Oncogene 2006;25:4683-96
  • Le Goffe C, Vallette G, Charrier L, et al. Metabolic control of resistance of human epithelial cells to H2O2 and NO stresses. Biochem J 2002;364:349-59
  • Kurtoglu M, Gao N, Shang J, et al. Under normoxia, 2-deoxy-D-glucose elicits cell death in select tumor types not by inhibition of glycolysis but by interfering with N-linked glycosylation. Mol Cancer Ther 2007;6:3049-58
  • Kang HT, Hwang ES. 2-Deoxyglucose: an anticancer and antiviral therapeutic, but not any more a low glucose mimetic. Life Sci 2006;78:1392-9
  • Liu H, Savaraj N, Priebe W, Lampidis TJ. Hypoxia increases tumor cell sensitivity to glycolytic inhibitors: a strategy for solid tumor therapy (Model C). Biochem Pharmacol 2002;64:1745-51
  • Maher JC, Krishan A, Lampidis TJ. Greater cell cycle inhibition and cytotoxicity induced by 2-deoxy-D-glucose in tumor cells treated under hypoxic vs aerobic conditions. Cancer Chemother Pharmacol 2004;53:116-22
  • Maschek G, Savaraj N, Priebe W, et al. 2-deoxy-D-glucose increases the efficacy of adriamycin and paclitaxel in human osteosarcoma and non-small cell lung cancers in vivo. Cancer Res 2004;64:31-4
  • Ledoux S, Yang R, Friedlander G, Laouari D. Glucose depletion enhances P-glycoprotein expression in hepatoma cells: role of endoplasmic reticulum stress response. Cancer Res 2003;63:7284-90
  • Mohanti BK, Rath GK, Anantha N, et al. Improving cancer radiotherapy with 2-deoxy-D-glucose: phase I/II clinical trials on human cerebral gliomas. Int J Radiat Oncol Biol Phys 1996;35:103-11
  • Singh D, Banerji AK, Dwarakanath BS, et al. Optimizing cancer radiotherapy with 2-deoxy-d-glucose dose escalation studies in patients with glioblastoma multiforme. Strahlenther Onkol 2005;181:507-14
  • Robey RB, Hong R, Zhong L, et al. Effects of the anti-tumor agent 3-bromopyruvate (3BrPA) on glycolytic energy metabolism. FASEB J 2007;21:890-6
  • Ko YH, Pedersen PL, Geschwind JF. Glucose catabolism in the rabbit VX2 tumor model for liver cancer: characterization and targeting hexokinase. Cancer Lett 2001;173:83-91
  • Geschwind JF, Ko YH, Torbenson MS, et al. Novel therapy for liver cancer: direct intraarterial injection of a potent inhibitor of ATP production. Cancer Res 2002;62:3909-13
  • Harrison SA, Di Bisceglie AM. A new approach to therapy for hepatocellular carcinoma? Hepatology 2003;37:471-3
  • Foubister V. Energy blocker to treat liver cancer. Drug Discov Today 2002;7:934-5
  • Ko YH, Smith BL, Wang Y, et al. Advanced cancers: eradication in all cases using 3-bromopyruvate therapy to deplete ATP. Biochem Biophys Res Commun 2004;324:269-75
  • Vali M, Liapi E, Kowalski J, et al. Intraarterial therapy with a new potent inhibitor of tumor metabolism (3-bromopyruvate): identification of therapeutic dose and method of injection in an animal model of liver cancer. J Vasc Interv Radiol 2007;18:95-101
  • Chang JM, Chung JW, Jae HJ, et al. Local toxicity of hepatic arterial infusion of hexokinase II inhibitor, 3-bromopyruvate: in vivo investigation in normal rabbit model. Acad Radiol 2007;14:85-92
  • Shin SW, Han H, Choo SW, et al. Hepatic intra-arterial injection of 3-bromopyruvate in rabbit VX2 tumor. Acta Radiol 2006;47:1036-41
  • Xu RH, Pelicano H, Zhou Y, et al. Inhibition of glycolysis in cancer cells: a novel strategy to overcome drug resistance associated with mitochondrial respiratory defect and hypoxia. Cancer Res 2005;65:613-21
  • Buijs M, Vossen JA, Geschwind JF, et al. Specificity of the anti-glycolytic activity of 3-bromopyruvate confirmed by FDG uptake in a rat model of breast cancer. Invest New Drugs 2008 [doi: 10.1007/s10637-008-9145-0]
  • Cao X, Bloomston M, Zhang T, et al. Synergistic antipancreatic tumor effect by simultaneously targeting hypoxic cancer cells with HSP90 inhibitor and glycolysis inhibitor. Clin Cancer Res 2008;14:1831-9
  • Meng F, Matteucci MD, Har CP. The antiproliferative activity of 3-bromopyruvate is not due to selective inhibition of glycolysis [abstract no. 2714]. In: Proceedings of the 99th Annual Meeting of the American Association for Cancer Research; 2008 Apr 12 – 16; San Diego, CA. Philadelphia (PA): AACR; 2008
  • Brawer MK. Lonidamine: basic science and rationale for treatment of prostatic proliferative disorders. Rev Urol 2005;7(Suppl 7):S21-6
  • Di Cosimo S, Ferretti G, Papaldo P, et al. Lonidamine: efficacy and safety in clinical trials for the treatment of solid tumors. Drugs Today (Barc) 2003;39:157-74
  • Sweetman SC, editor. Martindale: the complete drug reference. 35th edition. Electronic version. Pharmaceutical Press, London; 2005
  • Amadori D, Frassineti GL, De Matteis A, et al. Modulating effect of lonidamine on response to doxorubicin in metastatic breast cancer patients: results from a multicenter prospective randomized trial. Breast Cancer Res Treat 1998;49:209-17
  • De Lena M, Lorusso V, Latorre A, et al. Paclitaxel, cisplatin and lonidamine in advanced ovarian cancer. A phase II study. Eur J Cancer 2001;37:364-8
  • Oudard S, Carpentier A, Banu E, et al. Phase II study of lonidamine and diazepam in the treatment of recurrent glioblastoma multiforme. J Neurooncol 2003;63:81-6
  • Oudard S, Miccoli L, Dutrillaux B, Poupon MF. Targeting the gene of glucose metabolism for the treatment of advanced gliomas. Bull Cancer 1998;85:622-6
  • Miccoli L, Poirson-Bichat F, Sureau F, et al. Potentiation of lonidamine and diazepam, two agents acting on mitochondria, in human glioblastoma treatment. J Natl Cancer Inst 1998;90:1400-6
  • Sun AQ, Yüksel KU, Jacobson TM, Gracy RW. Isolation and characterization of human glucose-6-phosphate isomerase isoforms containing two different size subunits. Arch Biochem Biophys 1990;283:120-31
  • Funasaka T, Hu H, Hogan V, Raz A. Down-regulation of phosphoglucose isomerase/autocrine motility factor expression sensitizes human fibrosarcoma cells to oxidative stress leading to cellular senescence. J Biol Chem 2007;282:36362-9
  • Funasaka T, Hu H, Yanagawa T, et al. Down-regulation of phosphoglucose isomerase/autocrine motility factor results in mesenchymal-to-epithelial transition of human lung fibrosarcoma cells. Cancer Res 2007;67:4236-43
  • Haga A, Tanaka N, Funasaka T, et al. The autocrine motility factor (AMF) and AMF-receptor combination needs sugar chain recognition ability and interaction using the C-terminal region of AMF. J Mol Biol 2006;358:741-53
  • Tanaka N, Haga A, Naba N, et al. Crystal structures of mouse autocrine motility factor in complex with carbohydrate phosphate inhibitors provide insight into structure-activity relationship of the inhibitors. J Mol Biol 2006;356:312-24
  • Rathmell JC, Fox CJ, Plas DR, et al. Akt-directed glucose metabolism can prevent Bax conformation change and promote growth factor-independent survival. Mol Cell Biol 2003;23:7315-28
  • Bando H, Atsumi T, Nishio T, et al. Phosphorylation of the 6-phosphofructo-2-kinase/fructose2,6-bisphosphatase/PFKFB3 family of glycolytic regulators in human cancer. Clin Cancer Res 2005;11:5784-92
  • Chesney J. 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase and tumor cell glycolysis. Curr Opin Clin Nutr Metab Care 2006;9:535-9
  • 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 2008;7:110-20
  • Marin-Hernandez A, Rodríguez-Enríquez S, Vital-González PA, et al. Determining and understanding the control of glycolysis in fast-growth tumor cells. Flux control by an over-expressed but strongly product-inhibited hexokinase. FEBS J 2006;273:1975-88
  • Peng SY, Lai PL, Pan HW, et al. Aberrant expression of the glycolytic enzymes aldolase B and type II hexokinase in hepatocellular carcinoma are predictive markers for advanced stage, early recurrence and poor prognosis. Oncol Rep 2008;19:1045-53
  • Old SE, Landa LE, Mohrenweiser HW. Hominoid triosephosphate isomerase: regulation of expression of the proliferation specific isozyme. Mol Cell Biochem 1989;89:73-85
  • Ismail SA, Park HW. Structural analysis of human liver glyceraldehyde-3-phosphate dehydrogenase. Acta Crystallogr D Biol Crystallogr 2005;61:1508-13
  • Lin SJ, Guarente L. Nicotinamide adenine dinucleotide, a metabolic regulator of transcription, longevity and disease. Curr Opin Cell Biol 2003;15:241-6
  • Sundararaj KP, Wood RE, Ponnusamy S, et al. Rapid shortening of telomere length in response to ceramide involves the inhibition of telomere binding activity of nuclear glyceraldehyde-3-phosphate dehydrogenase. J Biol Chem 2004;279:6152-62
  • Colell A, Ricci JE, Tait S, et al. GAPDH and autophagy preserve survival after apoptotic cytochrome c release in the absence of caspase activation. Cell 2007;129:983-97
  • Song S, Finkel T. GAPDH and the search for alternative energy. Nat Cell Biol 2007;9:869-70
  • Saunders PA, Chen RW, Chuang DM. Nuclear translocation of glyceraldehyde-3-phosphate dehydrogenase isoforms during neuronal apoptosis. J Neurochem 1999;72:925-32
  • Bressi JC, Verlinde CL, Aronov AM, et al. Adenosine analogues as selective inhibitors of glyceraldehyde-3-phosphate dehydrogenase of Trypanosomatidae via structure-based drug design. J Med Chem 2001;44:2080-93
  • Suresh S, Bressi JC, Kennedy KJ, et al. Conformational changes in Leishmania mexicana glyceraldehyde-3-phosphate dehydrogenase induced by designed inhibitors. J Mol Biol 2001;309:423-35
  • Lai1 JCK, Bhardwaj V, Rizvi N, et al. Glycolytic enzyme inhibitors as novel anti-cancer drugs. The Northwest Regional Meeting; 2007 June 17 – 20; Boise, ID
  • Rodríguez-Enríquez S, Vital-González PA, Flores-Rodríguez FL, et al. Control of cellular proliferation by modulation of oxidative phosphorylation in human and rodent fast-growing tumor cells. Toxicol Appl Pharmacol 2006;215:208-17
  • Paoluzzi L, Gonen M, Gardner JR, et al. Targeting Bcl-2 family members with the BH3 mimetic AT-101 markedly enhances the therapeutic effects of chemotherapeutic agents in in vitro and in vivo models of B-cell lymphoma. Blood 2008;111:5350-8
  • Yoshioka H, Geyer CB, Hornecker JL, et al. In vivo analysis of developmentally and evolutionarily dynamic protein-DNA interactions regulating transcription of the Pgk2 gene during mammalian spermatogenesis. Mol Cell Biol 2007;27:7871-85
  • Duan Z, Lamendola DE, Yusuf RZ, et al. Overexpression of human phosphoglycerate kinase 1 (PGK1) induces a multi-drug resistance phenotype. Anticancer Res 2002;22:1933-41
  • Chen G, Gharib TG, Wang H, et al. Protein profiles associated with survival in lung adenocarcinoma. Proc Natl Acad Sci USA 2003;100:13537-42
  • Narasimhan K, Changqing Z, Choolani M. Ovarian cancer proteomics: Many technologies one goal. Proteomics Clin Appl 2008;2:195-218
  • Thierolf M, Hagmann ML, Pfeffer M, et al. Towards a comprehensive proteome of normal and malignant human colon tissue by 2-D-LC-ESI-MS and 2-DE proteomics and identification of S100A12 as potential cancer biomarker Proteomics Clin Appl 2008;2:11-22
  • Lay AJ, Jiang XM, Kisker O, et al. Phosphoglycerate kinase acts in tumour angiogenesis as a disulphide reductase. Nature 2000;408:869-73
  • Daly EB, Wind T, Jiang XM, et al. Secretion of phosphoglycerate kinase from tumour cells is controlled by oxygen-sensing hydroxylases. Biochim Biophys Acta 2004;1691:17-22
  • Fothergill LA, Herman C, Watson TI. The phosphoglycerate mutases. Adv Enzymol Mol Biol 2006;62:227-13
  • Li C, Xiao Z, Chen Z, et al. Proteome analysis of human lung squamous carcinoma. Proteomics 2006;6:547-58
  • Scatena R, Bottoni P, Giardina B, et al. Modulation of cancer cell line differentiation: a neglected proteomic analysis with potential implications in pathophysiology, diagnosis, prognosis, and therapy of cancer. Proteomics Clin Appl 2008;2:229-37
  • Kondoh H, Lleonart ME, Gil J, et al. Glycolytic enzymes can modulate cellular life span. Cancer Res 2005;65:177-85
  • Pancholi V. Multifunctional alpha-enolase: its role in diseases. Cell Mol Life Sci 2001;58:902-20
  • Mazurek S, Boschek CB, Hugo F, Eigenbrodt E. Pyruvate kinase type M2 and its role in tumor growth and spreading. Semin Cancer Biol 2005;15:300-8
  • García-Alles LF, Erni B. Synthesis of phosphoenol pyruvate (PEP) analogues and evaluation as inhibitors of PEP-utilizing enzymes. Eur J Biochem 2002;269:3226-36
  • Overington JP, Al-Lazikani B, Hopkins AL. How many drug targets are there? Nat Rev Drug Discov 2006;5:993-6
  • Kolev Y, Uetake H, Takagi Y, Sugihara K. Lactate dehydrogenase-5 (LDH-5) expression in human gastric cancer: association with hypoxia-inducible factor (HIF-1alpha) pathway, angiogenic factors production and poor prognosis. Ann Surg Oncol 2008;15:2336-44
  • Choueiri TK. Factors associated with outcome in patients with advanced renal cell carcinoma in the era of antiangiogenic agents. Clin Genitourin Cancer 2008;6:15-20
  • Wimazal F, Sperr WR, Kundi M, et al. Prognostic significance of serial determinations of lactate dehydrogenase (LDH) in the follow-up of patients with myelodysplastic syndromes. Ann Oncol 2008;19:970-6
  • George S, Bukowski RM. Biomarkers in clear cell renal cell carcinoma. Expert Rev Anticancer Ther 2007;7:1737-47
  • Fantin VR, St-Pierre J, Leder P. Attenuation of LDH-A expression uncovers a link between glycolysis, mitochondrial physiology, and tumor maintenance. Cancer Cell 2006;9:425-34
  • Choi SR, Pradhan A, Hammond NL, et al. Design, synthesis, and biological evaluation of Plasmodium falciparum lactate dehydrogenase inhibitors. J Med Chem 2007;50:3841-50
  • Deck LM, Royer RE, Chamblee BB, et al. Selective inhibitors of human lactate dehydrogenases and lactate dehydrogenase from the malarial parasite Plasmodium falciparum. J Med Chem 1998;41:3879-87

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.