Brain Tumor Res Treat.  2013 Oct;1(2):57-63. 10.14791/btrt.2013.1.2.57.

Pyruvate Dehydrogenase Kinase as a Potential Therapeutic Target for Malignant Gliomas

Affiliations
  • 1Department of Pharmacology, Brain Science & Engineering Institute, Kyungpook National University School of Medicine, Daegu, Korea. ksuk@knu.ac.kr

Abstract

Metabolic aberrations in the form of altered flux through key metabolic pathways are the major hallmarks of several life-threatening malignancies including malignant gliomas. These adaptations play an important role in the enhancement of the survival and proliferation of gliomas at the expense of the surrounding normal/healthy tissues. Recent studies in the field of neurooncology have directly targeted the altered metabolic pathways of malignant tumor cells for the development of anti-cancer drugs. Aerobic glycolysis due to elevated production of lactate from pyruvate regardless of oxygen availability is a common metabolic alteration in most malignancies. Aerobic glycolysis offers survival advantages in addition to generating substrates such as fatty acids, amino acids and nucleotides required for the rapid proliferation of cells. This review outlines the role of pyruvate dehydrogenase kinase (PDK) in gliomas as an inhibitor of pyruvate dehydrogenase that catalyzes the oxidative decarboxylation of pyruvate. An in-depth investigation on the key metabolic enzyme PDK may provide a novel therapeutic approach for the treatment of malignant gliomas.

Keyword

Pyruvate dehydrogenase kinase; Glioma; Gliomagenesis; Dichloroacetate; Hypoxia-inducible factor

MeSH Terms

Amino Acids
Decarboxylation
Dichloroacetic Acid
Fatty Acids
Glioma*
Glycolysis
Lactic Acid
Metabolic Networks and Pathways
Nucleotides
Oxidoreductases*
Oxygen
Phosphotransferases*
Pyruvic Acid*
Amino Acids
Fatty Acids
Lactic Acid
Nucleotides
Oxidoreductases
Oxygen
Phosphotransferases
Pyruvic Acid

Figure

  • Fig. 1 Classification of gliomas based on the origin of the cells. Adult neural stem cells and multipotent progenitors differentiate into mature neurons, astrocytes and oligodendrocytes. Neural stem cells and different types of progenitor cells can form gliomas. Mature astrocytes and oligodendrocytes also serve as cells of origin for gliomas.

  • Fig. 2 Potential role played by PDKs in malignant gliomas. Glucose entering the cell is metabolized by glycolysis to pyruvate. Most of the pyruvate in non-cancerous cells enters the mitochondria under aerobic conditions and a small fraction is metabolized to lactate. PDH in mitochondria converts pyruvate into acetyl-CoA, which enters the TCA cycle. On the other hand, in glioma cells, the oxidative (mitochondrial) pathway of glucose utilization is suppressed, and most of the pyruvate is converted into lactate. Hence, the relative activities of LDH and PDH determine the fate of pyruvate. PDK, which can be inhibited by DCA, inhibits PDH through phosphorylation and regulates its activities. HIF-1 induces PDK, which inactivates PDH resulting in suppression of the TCA cycle and mitochondrial respiration. Moreover, HIF-1 also stimulates glycolysis and the expression of LDH, thereby facilitating the conversion of pyruvate into lactate. DCA: dichloroacetate, HIF: hypoxia-inducible factor, LDH: lactate dehydrogenase, PDH: pyruvate dehydrogenase, PDK: pyruvate dehydrogenase kinase, TCA: tricarboxylic acid.


Reference

1. Jha MK, Jeon S, Suk K. Pyruvate Dehydrogenase Kinases in the Nervous System: Their Principal Functions in Neuronal-glial Metabolic Interaction and Neuro-metabolic Disorders. Curr Neuropharmacol. 2012; 10:393–403.
Article
2. Patel MS, Roche TE. Molecular biology and biochemistry of pyruvate dehydrogenase complexes. FASEB J. 1990; 4:3224–3233.
3. Mamelak AN, Jacoby DB. Targeted delivery of antitumoral therapy to glioma and other malignancies with synthetic chlorotoxin (TM-601). Expert Opin Drug Deliv. 2007; 4:175–186.
Article
4. Louis DN. Molecular pathology of malignant gliomas. Annu Rev Pathol. 2006; 1:97–117.
Article
5. Ricard D, Idbaih A, Ducray F, Lahutte M, Hoang-Xuan K, Delattre JY. Primary brain tumours in adults. Lancet. 2012; 379:1984–1996.
Article
6. Galli R, Binda E, Orfanelli U, et al. Isolation and characterization of tumorigenic, stem-like neural precursors from human glioblastoma. Cancer Res. 2004; 64:7011–7021.
Article
7. Singh SK, Hawkins C, Clarke ID, et al. Identification of human brain tumour initiating cells. Nature. 2004; 432:396–401.
Article
8. Goodenberger ML, Jenkins RB. Genetics of adult glioma. Cancer Genet. 2012; 205:613–621.
Article
9. Dolecek TA, Propp JM, Stroup NE, Kruchko C. CBTRUS statistical report: primary brain and central nervous system tumors diagnosed in the United States in 2005-2009. Neuro Oncol. 2012; 14:Suppl 5. v1–v49.
Article
10. Patil SA, Hosni-Ahmed A, Jones TS, Patil R, Pfeffer LM, Miller DD. Novel approaches to glioma drug design and drug screening. Expert Opin Drug Discov. 2013; 8:1135–1151.
Article
11. Brat DJ, Mapstone TB. Malignant glioma physiology: cellular response to hypoxia and its role in tumor progression. Ann Intern Med. 2003; 138:659–668.
Article
12. Brat DJ, Van Meir EG. Vaso-occlusive and prothrombotic mechanisms associated with tumor hypoxia, necrosis, and accelerated growth in glioblastoma. Lab Invest. 2004; 84:397–405.
Article
13. Brat DJ, Castellano-Sanchez AA, Hunter SB, et al. Pseudopalisades in glioblastoma are hypoxic, express extracellular matrix proteases, and are formed by an actively migrating cell population. Cancer Res. 2004; 64:920–927.
Article
14. Maxwell PH, Pugh CW, Ratcliffe PJ. Activation of the HIF pathway in cancer. Curr Opin Genet Dev. 2001; 11:293–299.
Article
15. Seyfried TN, Shelton LM. Cancer as a metabolic disease. Nutr Metab (Lond). 2010; 7:7.
Article
16. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011; 144:646–674.
Article
17. Abbott RG, Forrest S, Pienta KJ. Simulating the hallmarks of cancer. Artif Life. 2006; 12:617–634.
Article
18. Cavallo F, De Giovanni C, Nanni P, Forni G, Lollini PL. 2011: the immune hallmarks of cancer. Cancer Immunol Immunother. 2011; 60:319–326.
Article
19. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000; 100:57–70.
Article
20. Seyfried TN, Mukherjee P. Targeting energy metabolism in brain cancer: review and hypothesis. Nutr Metab (Lond). 2005; 2:30.
Article
21. Semenza GL, Artemov D, Bedi A, et al. 'The metabolism of tumours': 70 years later. Novartis Found Symp. 2001; 240:251–260. discussion 260-4.
Article
22. Ristow M. Oxidative metabolism in cancer growth. Curr Opin Clin Nutr Metab Care. 2006; 9:339–345.
Article
23. Gatenby RA, Gillies RJ. Why do cancers have high aerobic glycolysis? Nat Rev Cancer. 2004; 4:891–899.
Article
24. Gogvadze V, Orrenius S, Zhivotovsky B. Mitochondria in cancer cells: what is so special about them? Trends Cell Biol. 2008; 18:165–173.
Article
25. Maurer GD, Brucker DP, Bähr O, et al. Differential utilization of ketone bodies by neurons and glioma cell lines: a rationale for ketogenic diet as experimental glioma therapy. BMC Cancer. 2011; 11:315.
Article
26. Harris AL. Hypoxia--a key regulatory factor in tumour growth. Nat Rev Cancer. 2002; 2:38–47.
27. Provenzano PP, Cuevas C, Chang AE, Goel VK, Von Hoff DD, Hingorani SR. Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell. 2012; 21:418–429.
Article
28. Bertout JA, Patel SA, Simon MC. The impact of O2 availability on human cancer. Nat Rev Cancer. 2008; 8:967–975.
Article
29. Semenza GL. Oxygen sensing, homeostasis, and disease. N Engl J Med. 2011; 365:537–547.
Article
30. Goel S, Duda DG, Xu L, et al. Normalization of the vasculature for treatment of cancer and other diseases. Physiol Rev. 2011; 91:1071–1121.
Article
31. Semenza GL. Hypoxia-inducible factors in physiology and medicine. Cell. 2012; 148:399–408.
Article
32. Sutendra G, Michelakis ED. Pyruvate dehydrogenase kinase as a novel therapeutic target in oncology. Front Oncol. 2013; 3:38.
Article
33. Warburg O. On the origin of cancer cells. Science. 1956; 123:309–314.
Article
34. Warburg O. On respiratory impairment in cancer cells. Science. 1956; 124:269–270.
Article
35. Kim JW, Dang CV. Cancer's molecular sweet tooth and the Warburg effect. Cancer Res. 2006; 66:8927–8930.
Article
36. Bertram JS. The molecular biology of cancer. Mol Aspects Med. 2000; 21:167–223.
Article
37. Grandér D. How do mutated oncogenes and tumor suppressor genes cause cancer? Med Oncol. 1998; 15:20–26.
Article
38. Griguer CE, Oliva CR, Gillespie GY. Glucose metabolism heterogeneity in human and mouse malignant glioma cell lines. J Neurooncol. 2005; 74:123–133.
Article
39. Morfouace M, Lalier L, Bahut M, et al. Comparison of spheroids formed by rat glioma stem cells and neural stem cells reveals differences in glucose metabolism and promising therapeutic applications. J Biol Chem. 2012; 287:33664–33674.
Article
40. van Horssen R, Willemse M, Haeger A, et al. Intracellular NAD(H) levels control motility and invasion of glioma cells. Cell Mol Life Sci. 2013; 70:2175–2190.
Article
41. Brizel DM, Schroeder T, Scher RL, et al. Elevated tumor lactate concentrations predict for an increased risk of metastases in head-and-neck cancer. Int J Radiat Oncol Biol Phys. 2001; 51:349–353.
Article
42. Cairns RA, Bennewith KL, Graves EE, Giaccia AJ, Chang DT, Denko NC. Pharmacologically increased tumor hypoxia can be measured by 18F-Fluoroazomycin arabinoside positron emission tomography and enhances tumor response to hypoxic cytotoxin PR-104. Clin Cancer Res. 2009; 15:7170–7174.
Article
43. Zwicker F, Kirsner A, Peschke P, et al. Dichloroacetate induces tumor-specific radiosensitivity in vitro but attenuates radiation-induced tumor growth delay in vivo. Strahlenther Onkol. 2013; 189:684–692.
Article
44. Ralph SJ, Rodríguez-Enríquez S, Neuzil J, Moreno-Sánchez R. Bioenergetic pathways in tumor mitochondria as targets for cancer therapy and the importance of the ROS-induced apoptotic trigger. Mol Aspects Med. 2010; 31:29–59.
Article
45. Oudard S, Arvelo F, Miccoli L, et al. High glycolysis in gliomas despite low hexokinase transcription and activity correlated to chromosome 10 loss. Br J Cancer. 1996; 74:839–845.
Article
46. Mineura K, Yasuda T, Kowada M, Shishido F, Ogawa T, Uemura K. Positron emission tomographic evaluation of histological malignancy in gliomas using oxygen-15 and fluorine-18-fluorodeoxyglucose. Neurol Res. 1986; 8:164–168.
Article
47. Roche TE, Hiromasa Y. Pyruvate dehydrogenase kinase regulatory mechanisms and inhibition in treating diabetes, heart ischemia, and cancer. Cell Mol Life Sci. 2007; 64:830–849.
Article
48. Ke Q, Costa M. Hypoxia-inducible factor-1 (HIF-1). Mol Pharmacol. 2006; 70:1469–1480.
Article
49. Wu G, Luo J, Rana JS, Laham R, Sellke FW, Li J. Involvement of COX-2 in VEGF-induced angiogenesis via P38 and JNK pathways in vascular endothelial cells. Cardiovasc Res. 2006; 69:512–519.
Article
50. Airley RE, Mobasheri A. Hypoxic regulation of glucose transport, anaerobic metabolism and angiogenesis in cancer: novel pathways and targets for anticancer therapeutics. Chemotherapy. 2007; 53:233–256.
Article
51. Liang C, Guo S, Yang L. All-trans retinoic acid upregulates VEGF expression in glioma cells in vitro. J Biomed Res. 2013; 27:51–55.
Article
52. Greer SN, Metcalf JL, Wang Y, Ohh M. The updated biology of hypoxia-inducible factor. EMBO J. 2012; 31:2448–2460.
Article
53. Pescador N, Villar D, Cifuentes D, et al. Hypoxia promotes glycogen accumulation through hypoxia inducible factor (HIF)-mediated induction of glycogen synthase 1. PLoS One. 2010; 5:e9644.
Article
54. Tong JJ, Yan Z, Jian R, Tao H, Hui OT, Jian C. RhoA regulates invasion of glioma cells via the c-Jun NH2-terminal kinase pathway under hypoxia. Oncol Lett. 2012; 4:495–500.
Article
55. 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. 2006; 3:177–185.
Article
56. Walenta S, Snyder S, Haroon ZA, et al. Tissue gradients of energy metabolites mirror oxygen tension gradients in a rat mammary carcinoma model. Int J Radiat Oncol Biol Phys. 2001; 51:840–848.
Article
57. 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 Cell. 2007; 11:37–51.
Article
58. Knoechel TR, Tucker AD, Robinson CM, et al. Regulatory roles of the N-terminal domain based on crystal structures of human pyruvate dehydrogenase kinase 2 containing physiological and synthetic ligands. Biochemistry. 2006; 45:402–415.
Article
59. Bowker-Kinley MM, Davis WI, Wu P, Harris RA, Popov KM. Evidence for existence of tissue-specific regulation of the mammalian pyruvate dehydrogenase complex. Biochem J. 1998; 329(Pt 1):191–196.
Article
60. Baker JC, Yan X, Peng T, Kasten S, Roche TE. Marked differences between two isoforms of human pyruvate dehydrogenase kinase. J Biol Chem. 2000; 275:15773–15781.
Article
61. Wong JY, Huggins GS, Debidda M, Munshi NC, De Vivo I. Dichloroacetate induces apoptosis in endometrial cancer cells. Gynecol Oncol. 2008; 109:394–402.
Article
62. Stacpoole PW, Nagaraja NV, Hutson AD. Efficacy of dichloroacetate as a lactate-lowering drug. J Clin Pharmacol. 2003; 43:683–691.
Article
63. Michelakis ED, Webster L, Mackey JR. Dichloroacetate (DCA) as a potential metabolic-targeting therapy for cancer. Br J Cancer. 2008; 99:989–994.
Article
64. Pearson H. Cancer patients opt for unapproved drug. Nature. 2007; 446:474–475.
Article
65. Papandreou I, Goliasova T, Denko NC. Anticancer drugs that target metabolism: is dichloroacetate the new paradigm? Int J Cancer. 2011; 128:1001–1008.
Article
66. Yaromina A, Meyer S, Fabian C, et al. Effects of three modifiers of glycolysis on ATP, lactate, hypoxia, and growth in human tumor cell lines in vivo. Strahlenther Onkol. 2012; 188:431–437.
Article
67. Stockwin LH, Yu SX, Borgel S, et al. Sodium dichloroacetate selectively targets cells with defects in the mitochondrial ETC. Int J Cancer. 2010; 127:2510–2519.
Article
68. Ralph SJ, Neuzil J. Mitochondria as targets for cancer therapy. Mol Nutr Food Res. 2009; 53:9–28.
Article
69. Cai P, Boor PJ, Khan MF, Kaphalia BS, Ansari GA, Konig R. Immuno- and hepato-toxicity of dichloroacetic acid in MRL(+/+) and B(6)C(3) F(1) mice. J Immunotoxicol. 2007; 4:107–115.
70. Michelakis ED, Sutendra G, Dromparis P, et al. Metabolic modulation of glioblastoma with dichloroacetate. Sci Transl Med. 2010; 2:31ra34.
Article
71. Huse JT, Holland EC. Targeting brain cancer: advances in the molecular pathology of malignant glioma and medulloblastoma. Nat Rev Cancer. 2010; 10:319–331.
Article
72. Duan Y, Zhao X, Ren W, et al. Antitumor activity of dichloroacetate on C6 glioma cell: in vitro and in vivo evaluation. Onco Targets Ther. 2013; 6:189–198.
73. Ferriero R, Manco G, Lamantea E, et al. Phenylbutyrate therapy for pyruvate dehydrogenase complex deficiency and lactic acidosis. Sci Transl Med. 2013; 5:175ra31.
Article
74. Bolden JE, Peart MJ, Johnstone RW. Anticancer activities of histone deacetylase inhibitors. Nat Rev Drug Discov. 2006; 5:769–784.
Article
Full Text Links
  • BTRT
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr