Skip to main content

Advertisement

Log in

Provocative Question: Should Ketogenic Metabolic Therapy Become the Standard of Care for Glioblastoma?

  • Original Paper
  • Published:
Neurochemical Research Aims and scope Submit manuscript

Abstract

No major advances have been made in improving overall survival for glioblastoma (GBM) in almost 100 years. The current standard of care (SOC) for GBM involves immediate surgical resection followed by radiotherapy with concomitant temozolomide chemotherapy. Corticosteroid (dexamethasone) is often prescribed to GBM patients to reduce tumor edema and inflammation. The SOC disrupts the glutamate–glutamine cycle thus increasing availability of glucose and glutamine in the tumor microenvironment. Glucose and glutamine are the prime fermentable fuels that underlie therapy resistance and drive GBM growth through substrate level phosphorylation in the cytoplasm and the mitochondria, respectively. Emerging evidence indicates that ketogenic metabolic therapy (KMT) can reduce glucose availability while elevating ketone bodies that are neuroprotective and non-fermentable. Information is presented from preclinical and case report studies showing how KMT could target tumor cells without causing neurochemical damage thus improving progression free and overall survival for patients with GBM.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

Abbreviations

GBM:

Glioblastoma

TMZ:

Temozolomide

SOC:

Standard of care

KMT:

Ketogenic metabolic therapy

References

  1. Polivka J Jr, Polivka J, Holubec L, Kubikova T, Priban V, Hes O, Pivovarcikova K, Treskova I (2017) Advances in experimental targeted therapy and immunotherapy for patients with glioblastoma multiforme. Anticancer Res 37:21–33

    Article  CAS  PubMed  Google Scholar 

  2. Fabbro-Peray P, Zouaoui S, Darlix A, Fabbro M, Pallud J, Rigau V, Mathieu-Daude H, Bessaoud F, Bauchet F, Riondel A, Sorbets E, Charissoux M, Amelot A, Mandonnet E, Figarella-Branger D, Duffau H, Tretarre B, Taillandier L, Bauchet L (2018) Association of patterns of care, prognostic factors, and use of radiotherapy-temozolomide therapy with survival in patients with newly diagnosed glioblastoma: a French national population-based study. J Neurooncol 142(1):91–101

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Geraldo LHM, Garcia C, da Fonseca ACC, Dubois LGF, de Sampaio ESTCL, Matias D, de Camargo Magalhaes ES, do Amaral RF, da Rosa BG, Grimaldi I, Leser FS, Janeiro JM, Macharia L, Wanjiru C, Pereira CM, Moura-Neto V, Freitas C, Lima FRS (2019) Glioblastoma therapy in the age of molecular medicine. Trends Cancer 5:46–65

    Article  CAS  PubMed  Google Scholar 

  4. Wegman-Ostrosky T, Reynoso-Noveron N, Mejia-Perez SI, Sanchez-Correa TE, Alvarez-Gomez RM, Vidal-Millan S, Cacho-Diaz B, Sanchez-Corona J, Herrera-Montalvo LA, Corona-Vazquez T (2016) Clinical prognostic factors in adults with astrocytoma: historic cohort. Clin Neurol Neurosurg 146:116–122

    Article  PubMed  Google Scholar 

  5. Chinopoulos C, Seyfried TN (2018) Mitochondrial substrate level phosphorylation as energy source for glioblastoma: review and hypothesis. ASN Neuro 10:1–27

    Article  Google Scholar 

  6. Fatehi M, Hunt C, Ma R, Toyota BD (2018) Persistent disparities in survival for patients with glioblastoma. World Neurosurg 120:e511–e516

    Article  PubMed  Google Scholar 

  7. Kleihues P, Ohgaki H (1999) Primary and secondary glioblastomas: from concept to clinical diagnosis. Neuro Oncol 1:44–51

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Scherer HJ (1940) A critical review: the pathology of cerebral gliomas. J Neurol Neuropsychiat 3:147–177

    Article  CAS  Google Scholar 

  9. Zagzag D, Esencay M, Mendez O, Yee H, Smirnova I, Huang Y, Chiriboga L, Lukyanov E, Liu M, Newcomb EW (2008) Hypoxia- and vascular endothelial growth factor-induced stromal cell-derived factor-1alpha/CXCR4 expression in glioblastomas: one plausible explanation of Scherer’s structures. Am J Pathol 173:545–560

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Shelton LM, Mukherjee P, Huysentruyt LC, Urits I, Rosenberg JA, Seyfried TN (2010) A novel pre-clinical in vivo mouse model for malignant brain tumor growth and invasion. J Neurooncol 99:165–176

    Article  CAS  PubMed  Google Scholar 

  11. Laws ER Jr, Goldberg WJ, Bernstein JJ (1993) Migration of human malignant astrocytoma cells in the mammalian brain: Scherer revisited. Int J Dev Neurosci 11:691–697

    Article  PubMed  Google Scholar 

  12. Perry JR, Laperriere N, O’Callaghan CJ, Brandes AA, Menten J, Phillips C, Fay M, Nishikawa R, Cairncross JG, Roa W, Osoba D, Rossiter JP, Sahgal A, Hirte H, Laigle-Donadey F, Franceschi E, Chinot O, Golfinopoulos V, Fariselli L, Wick A, Feuvret L, Back M, Tills M, Winch C, Baumert BG, Wick W, Ding K, Mason WP, Trial I (2017) Short-course radiation plus temozolomide in elderly patients with glioblastoma. N Engl J Med 376:1027–1037

    Article  CAS  PubMed  Google Scholar 

  13. Krex D, Klink B, Hartmann C, von Deimling A, Pietsch T, Simon M, Sabel M, Steinbach JP, Heese O, Reifenberger G, Weller M, Schackert G (2007) Long-term survival with glioblastoma multiforme. Brain 130:2596–2606

    Article  PubMed  Google Scholar 

  14. Cuddapah VA, Robel S, Watkins S, Sontheimer H (2014) A neurocentric perspective on glioma invasion. Nat Rev Neurosci 15:455–465

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Chang SM, Parney IF, Huang W, Anderson FA Jr, Asher AL, Bernstein M, Lillehei KO, Brem H, Berger MS, Laws ER (2005) Patterns of care for adults with newly diagnosed malignant glioma. JAMA 293:557–564

    Article  CAS  PubMed  Google Scholar 

  16. Taphoorn MJ, Stupp R, Coens C, Osoba D, Kortmann R, van den Bent MJ, Mason W, Mirimanoff RO, Baumert BG, Eisenhauer E, Forsyth P, Bottomley A (2005) Health-related quality of life in patients with glioblastoma: a randomised controlled trial. Lancet Oncol 6:937–944

    Article  PubMed  Google Scholar 

  17. Flechl B, Ackerl M, Sax C, Dieckmann K, Crevenna R, Gaiger A, Widhalm G, Preusser M, Marosi C (2012) Neurocognitive and sociodemographic functioning of glioblastoma long-term survivors. J Neurooncol 109:331–339

    Article  PubMed  Google Scholar 

  18. Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJ, Janzer RC, Ludwin SK, Allgeier A, Fisher B, Belanger K, Hau P, Brandes AA, Gijtenbeek J, Marosi C, Vecht CJ, Mokhtari K, Wesseling P, Villa S, Eisenhauer E, Gorlia T, Weller M, Lacombe D, Cairncross JG, Mirimanoff RO (2009) Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol 10:459–466

    Article  CAS  PubMed  Google Scholar 

  19. Morantz RA, Wood GW, Foster M, Clark M, Gollahon K (1979) Macrophages in experimental and human brain tumors. Part 2: studies of the macrophage content of human brain tumors. J Neurosurg 50:305–311

    Article  CAS  PubMed  Google Scholar 

  20. Morantz RA, Wood GW, Foster M, Clark M, Gollahon K (1979) Macrophages in experimental and human brain tumors. Part 1: studies of the macrophage content of experimental rat brain tumors of varying immunogenicity. J Neurosurg 50:298–304

    Article  CAS  PubMed  Google Scholar 

  21. Karsy M, Gelbman M, Shah P, Balumbu O, Moy F, Arslan E (2012) Established and emerging variants of glioblastoma multiforme: review of morphological and molecular features. Folia Neuropathol 50:301–321

    Article  PubMed  Google Scholar 

  22. Huysentruyt LC, Akgoc Z, Seyfried TN (2011) Hypothesis: are neoplastic macrophages/microglia present in glioblastoma multiforme? ASN Neuro 3(4):0011

    Article  Google Scholar 

  23. Yuan X, Curtin J, Xiong Y, Liu G, Waschsmann-Hogiu S, Farkas DL, Black KL, Yu JS (2004) Isolation of cancer stem cells from adult glioblastoma multiforme. Oncogene 23:9392–9400

    Article  CAS  PubMed  Google Scholar 

  24. Rubinstein LJ (1972) Tumors of the central nervous system. Armed Forces Institute of Pathology, Washington, D.C.

    Google Scholar 

  25. Wood GW, Morantz RA (1979) Immunohistologic evaluation of the lymphoreticular infiltrate of human central nervous system tumors. J Natl Cancer Inst 62:485–491

    Article  CAS  PubMed  Google Scholar 

  26. Seyfried TN (2001) Perspectives on brain tumor formation involving macrophages, glia, and neural stem cells. Perspect Biol Med 44:263–282

    Article  CAS  PubMed  Google Scholar 

  27. Roggendorf W, Strupp S, Paulus W (1996) Distribution and characterization of microglia/macrophages in human brain tumors. Acta Neuropathol 92:288–293

    Article  CAS  PubMed  Google Scholar 

  28. Ordys BB, Launay S, Deighton RF, McCulloch J, Whittle IR (2010) The role of mitochondria in glioma pathophysiology. Mol Neurobiol 42:64–75

    Article  CAS  PubMed  Google Scholar 

  29. Oudard S, Boitier E, Miccoli L, Rousset S, Dutrillaux B, Poupon MF (1997) Gliomas are driven by glycolysis: putative roles of hexokinase, oxidative phosphorylation and mitochondrial ultrastructure. Anticancer Res 17:1903–1911

    CAS  PubMed  Google Scholar 

  30. Seyfried TN, Yu G, Maroon JC, D’Agostino DP (2017) Press-pulse: a novel therapeutic strategy for the metabolic management of cancer. Nutr Metab (Lond) 14:19

    Article  CAS  Google Scholar 

  31. Feichtinger RG, Weis S, Mayr JA, Zimmermann F, Geilberger R, Sperl W, Kofler B (2014) Alterations of oxidative phosphorylation complexes in astrocytomas. Glia 62:514–525

    Article  PubMed  Google Scholar 

  32. Arismendi-Morillo GJ, Castellano-Ramirez AV (2008) Ultrastructural mitochondrial pathology in human astrocytic tumors: potentials implications pro-therapeutics strategies. J Electron Microsc (Tokyo) 57:33–39

    Article  Google Scholar 

  33. Katsetos CD, Anni H, Draber P (2013) Mitochondrial dysfunction in gliomas. Semin Pediatr Neurol 20:216–227

    Article  PubMed  Google Scholar 

  34. Deighton RF, Le Bihan T, Martin SF, Gerth AM, McCulloch M, Edgar JM, Kerr LE, Whittle IR, McCulloch J (2014) Interactions among mitochondrial proteins altered in glioblastoma. J Neurooncol 118:247–256

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Scheithauer BW, Bruner JM (1987) The ultrastructural spectrum of astrocytic neoplasms. Ultrastruct Pathol 11:535–581

    Article  CAS  PubMed  Google Scholar 

  36. Sipe JC, Herman MM, Rubinstein LJ (1973) Electron microscopic observations on human glioblastomas and astrocytomas maintained in organ culture systems. Am J Pathol 73:589–606

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Arismendi-Morillo G, Castellano-Ramirez A, Seyfried TN (2017) Ultrastructural characterization of the Mitochondria-associated membranes abnormalities in human astrocytomas: functional and therapeutics implications. Ultrastruct Pathol 41:234–244

    Article  PubMed  Google Scholar 

  38. Kiebish MA, Han X, Cheng H, Chuang JH, Seyfried TN (2008) Cardiolipin and electron transport chain abnormalities in mouse brain tumor mitochondria: lipidomic evidence supporting the Warburg theory of cancer. J Lipid Res 49:2545–2556

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Claypool SM, Koehler CM (2012) The complexity of cardiolipin in health and disease. Trends Biochem Sci 37:32–41

    Article  CAS  PubMed  Google Scholar 

  40. Guntuku L, Naidu VG, Yerra VG (2016) Mitochondrial dysfunction in gliomas: pharmacotherapeutic potential of natural compounds. Curr Neuropharmacol 14:567–583

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Bartesaghi S, Graziano V, Galavotti S, Henriquez NV, Betts J, Saxena J, Minieri V, Deli A, Karlsson A, Martins LM, Capasso M, Nicotera P, Brandner S, De Laurenzi V, Salomoni P (2015) Inhibition of oxidative metabolism leads to p53 genetic inactivation and transformation in neural stem cells. Proc Natl Acad Sci USA 112:1059–1064

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Libby CJ, Tran AN, Scott SE, Griguer C, Hjelmeland AB (2018) The pro-tumorigenic effects of metabolic alterations in glioblastoma including brain tumor initiating cells. Biochem Biophys Acta 1869:175–188

    CAS  Google Scholar 

  43. Lehninger AL (1964) The mitochondrion: molecular basis of structure and function. W.A. Benjamin, INC., New York

    Google Scholar 

  44. Seyfried TN, Shelton LM (2010) Cancer as a metabolic disease. Nutr Metab (Lond) 7:7

    Article  CAS  Google Scholar 

  45. Seyfried TN, Flores RE, Poff AM, D’Agostino DP (2014) Cancer as a metabolic disease: implications for novel therapeutics. Carcinogenesis 35:515–527

    Article  CAS  PubMed  Google Scholar 

  46. Seyfried TN (2015) Cancer as a mitochondrial metabolic disease. Front Cell Dev Biol 3:43

    Article  PubMed  PubMed Central  Google Scholar 

  47. Soto AM, Sonnenschein C (2004) The somatic mutation theory of cancer: growing problems with the paradigm? BioEssays 26:1097–1107

    Article  CAS  PubMed  Google Scholar 

  48. Sonnenschein C, Soto AM (1999) The society of cells: cancer and the control of cell proliferation. Springer, New York

    Google Scholar 

  49. Sonnenschein C, Soto AM (2000) Somatic mutation theory of carcinogenesis: why it should be dropped and replaced. Mol Carcinog 29:205–211

    Article  CAS  PubMed  Google Scholar 

  50. Szent-Gyorgyi A (1977) The living state and cancer. Proc Natl Acad Sci USA 74:2844–2847

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Vander Heiden MG, Cantley LC, Thompson CB (2009) Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 324:1029–1033

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Tanaka K, Sasayama T, Irino Y, Takata K, Nagashima H, Satoh N, Kyotani K, Mizowaki T, Imahori T, Ejima Y, Masui K, Gini B, Yang H, Hosoda K, Sasaki R, Mischel PS, Kohmura E (2015) Compensatory glutamine metabolism promotes glioblastoma resistance to mTOR inhibitor treatment. J Clin Invest 125:1591–1602

    Article  PubMed  PubMed Central  Google Scholar 

  53. Yang C, Sudderth J, Dang T, Bachoo RG, McDonald JG, Deberardinis RJ (2009) Glioblastoma cells require glutamate dehydrogenase to survive impairments of glucose metabolism or Akt signaling. Cancer Res 69(20):7986–7993

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. DeBerardinis RJ, Mancuso A, Daikhin E, Nissim I, Yudkoff M, Wehrli S, Thompson CB (2007) Beyond aerobic glycolysis: transformed cells can engage in glutamine metabolism that exceeds the requirement for protein and nucleotide synthesis. Proc Natl Acad Sci USA 104:19345–19350

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Marquez J, Alonso FJ, Mates JM, Segura JA, Martin-Rufian M, Campos-Sandoval JA (2017) Glutamine addiction in gliomas. Neurochem Res 42:1735–1746

    Article  CAS  PubMed  Google Scholar 

  56. Dahlberg D, Struys EA, Jansen EE, Morkrid L, Midttun O, Hassel B (2017) Cyst fluid from cystic, malignant brain tumors: a reservoir of nutrients, including growth factor-like nutrients, for tumor cells. Neurosurgery 80:917–924

    Article  PubMed  Google Scholar 

  57. Jaworski DM, Namboodiri AM, Moffett JR (2016) Acetate as a metabolic and epigenetic modifier of cancer therapy. J Cell Biochem 117:574–588

    Article  CAS  PubMed  Google Scholar 

  58. Rhodes CG, Wise RJ, Gibbs JM, Frackowiak RS, Hatazawa J, Palmer AJ, Thomas DG, Jones T (1983) In vivo disturbance of the oxidative metabolism of glucose in human cerebral gliomas. Ann Neurol 14:614–626

    Article  CAS  PubMed  Google Scholar 

  59. Flavahan WA, Wu Q, Hitomi M, Rahim N, Kim Y, Sloan AE, Weil RJ, Nakano I, Sarkaria JN, Stringer BW, Day BW, Li M, Lathia JD, Rich JN, Hjelmeland AB (2013) Brain tumor initiating cells adapt to restricted nutrition through preferential glucose uptake. Nat Neurosci 16:1373–1382

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. DeBerardinis RJ, Cheng T (2010) Q’s next: the diverse functions of glutamine in metabolism, cell biology and cancer. Oncogene 29:313–324

    Article  CAS  PubMed  Google Scholar 

  61. Yang L, Venneti S, Nagrath D (2017) Glutaminolysis: a hallmark of cancer metabolism. Annu Rev Biomed Eng 19:163–194

    Article  CAS  PubMed  Google Scholar 

  62. Amores-Sanchez MI, Medina MA (1999) Glutamine, as a precursor of glutathione, and oxidative stress. Mol Genet Metab 67:100–105

    Article  CAS  PubMed  Google Scholar 

  63. Xu RH, Pelicano H, Zhou Y, Carew JS, Feng L, Bhalla KN, Keating MJ, Huang P (2005) Inhibition of glycolysis in cancer cells: a novel strategy to overcome drug resistance associated with mitochondrial respiratory defect and hypoxia. Can Res 65:613–621

    CAS  Google Scholar 

  64. Elsakka AMA, Bary MA, Abdelzaher E, Elnaggar M, Kalamian M, Mukherjee P, Seyfried TN (2018) Management of glioblastoma multiforme in a patient treated with ketogenic metabolic therapy and modified standard of care: a 24-month follow-up. Front Nutr 5:20

    Article  PubMed  PubMed Central  Google Scholar 

  65. Pitter KL, Tamagno I, Alikhanyan K, Hosni-Ahmed A, Pattwell SS, Donnola S, Dai C, Ozawa T, Chang M, Chan TA, Beal K, Bishop AJ, Barker CA, Jones TS, Hentschel B, Gorlia T, Schlegel U, Stupp R, Weller M, Holland EC, Hambardzumyan D (2016) Corticosteroids compromise survival in glioblastoma. Brain 139:1458–1471

    Article  PubMed  PubMed Central  Google Scholar 

  66. Wong ET, Lok E, Gautam S, Swanson KD (2015) Dexamethasone exerts profound immunologic interference on treatment efficacy for recurrent glioblastoma. Br J Cancer 113:232–241

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Lawrence YR, Blumenthal DT, Matceyevsky D, Kanner AA, Bokstein F, Corn BW (2011) Delayed initiation of radiotherapy for glioblastoma: how important is it to push to the front (or the back) of the line? J Neurooncol 105(1):1–7

    Article  CAS  PubMed  Google Scholar 

  68. Lawrence YR, Wang M, Dicker AP, Andrews D, Curran WJ Jr, Michalski JM, Souhami L, Yung WK, Mehta M (2011) Early toxicity predicts long-term survival in high-grade glioma. Br J Cancer 104:1365–1371

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Alieva M, Margarido AS, Wieles T, Abels ER, Colak B, Boquetale C, Jan Noordmans H, Snijders TJ, Broekman ML, van Rheenen J (2017) Preventing inflammation inhibits biopsy-mediated changes in tumor cell behavior. Sci Rep 7:7529

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Walter ND, Rice PL, Redente EF, Kauvar EF, Lemond L, Aly T, Wanebo K, Chan ED (2011) Wound healing after trauma may predispose to lung cancer metastasis: review of potential mechanisms. Am J Respir Cell Mol Biol 44:591–596

    Article  CAS  PubMed  Google Scholar 

  71. Duan C, Yang R, Yuan L, Engelbach JA, Tsien CI, Rich KM, Dahiya SM, Johanns TM, Ackerman JJH, Garbow JR (2019) Late effects of radiation prime the brain microenvironment for accelerated tumor growth. Int J Radiat Oncol Biol Phys 103:190–194

    Article  PubMed  Google Scholar 

  72. Rovlias A, Kotsou S (2000) The influence of hyperglycemia on neurological outcome in patients with severe head injury. Neurosurgery 46:335–342 discussion 342–333

    Article  CAS  PubMed  Google Scholar 

  73. Bergsneider M, Hovda DA, Shalmon E, Kelly DF, Vespa PM, Martin NA, Phelps ME, McArthur DL, Caron MJ, Kraus JF, Becker DP (1997) Cerebral hyperglycolysis following severe traumatic brain injury in humans: a positron emission tomography study. J Neurosurg 86:241–251

    Article  CAS  PubMed  Google Scholar 

  74. Derr RL, Ye X, Islas MU, Desideri S, Saudek CD, Grossman SA (2009) Association between hyperglycemia and survival in patients with newly diagnosed glioblastoma. J Clin Oncol 27:1082–1086

    Article  PubMed  PubMed Central  Google Scholar 

  75. Mayer A, Vaupel P, Struss HG, Giese A, Stockinger M, Schmidberger H (2014) Strong adverse prognostic impact of hyperglycemic episodes during adjuvant chemoradiotherapy of glioblastoma multiforme. Strahlenther Onkol 190:933–938

    Article  PubMed  Google Scholar 

  76. McGirt MJ, Chaichana KL, Gathinji M, Attenello F, Than K, Ruiz AJ, Olivi A, Quinones-Hinojosa A (2008) Persistent outpatient hyperglycemia is independently associated with decreased survival after primary resection of malignant brain astrocytomas. Neurosurgery 63:286–291 discussion 291

    Article  PubMed  Google Scholar 

  77. Schwartzbaum J, Edlinger M, Zigmont V, Stattin P, Rempala GA, Nagel G, Hammar N, Ulmer H, Foger B, Walldius G, Manjer J, Malmstrom H, Feychting M (2017) Associations between prediagnostic blood glucose levels, diabetes, and glioma. Sci Rep 7:1436

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. Strowd RE 3rd, Grossman SA (2015) The role of glucose modulation and dietary supplementation in patients with central nervous system tumors. Curr Treat Options Oncol 16:356

    Article  Google Scholar 

  79. Tieu MT, Lovblom LE, McNamara MG, Mason W, Laperriere N, Millar BA, Menard C, Kiehl TR, Perkins BA, Chung C (2015) Impact of glycemia on survival of glioblastoma patients treated with radiation and temozolomide. J Neurooncol 124:119–126

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Zhao S, Cai J, Li J, Bao G, Li D, Li Y, Zhai X, Jiang C, Fan L (2016) Bioinformatic profiling identifies a glucose-related risk signature for the malignancy of glioma and the survival of patients. Mol Neurobiol

  81. Decker M, Sacks P, Abbatematteo J, De Leo E, Brennan M, Rahman M (2019) The effects of hyperglycemia on outcomes in surgical high-grade glioma patients. Clin Neurol Neurosurg 179:9–13

    Article  PubMed  Google Scholar 

  82. Link TW, Woodworth GF, Chaichana KL, Grossman SA, Mayer RS, Brem H, Weingart JD, Quinones-Hinojosa A (2012) Hyperglycemia is independently associated with post-operative function loss in patients with primary eloquent glioblastoma. J Clin Neurosci 19:996–1000

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Davies PS, Powell AE, Swain JR, Wong MH (2009) Inflammation and proliferation act together to mediate intestinal cell fusion. PLoS ONE 4:e6530

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  84. Seyfried TN, Huysentruyt LC (2013) On the origin of cancer metastasis. Crit Rev Oncog 18:43–73

    Article  PubMed  PubMed Central  Google Scholar 

  85. Pawelek JM, Chakraborty AK (2008) Fusion of tumour cells with bone marrow-derived cells: a unifying explanation for metastasis. Nat Rev Cancer 8:377–386

    Article  CAS  PubMed  Google Scholar 

  86. Lindstrom A, Midtbo K, Arnesson LG, Garvin S, Shabo I (2017) Fusion between M2-macrophages and cancer cells results in a subpopulation of radioresistant cells with enhanced DNA-repair capacity. Oncotarget 8:51370–51386

    Article  PubMed  PubMed Central  Google Scholar 

  87. Tardito S, Oudin A, Ahmed SU, Fack F, Keunen O, Zheng L, Miletic H, Sakariassen PO, Weinstock A, Wagner A, Lindsay SL, Hock AK, Barnett SC, Ruppin E, Morkve SH, Lund-Johansen M, Chalmers AJ, Bjerkvig R, Niclou SP, Gottlieb E (2015) Glutamine synthetase activity fuels nucleotide biosynthesis and supports growth of glutamine-restricted glioblastoma. Nat Cell Biol 17:1556–1568

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Takano T, Lin JH, Arcuino G, Gao Q, Yang J, Nedergaard M (2001) Glutamate release promotes growth of malignant gliomas. Nat Med 7:1010–1015

    Article  CAS  PubMed  Google Scholar 

  89. Seyfried TN, Shelton LM, Mukherjee P (2010) Does the existing standard of care increase glioblastoma energy metabolism? Lancet Oncol 11:811–813

    Article  PubMed  Google Scholar 

  90. Seyfried TN, Flores R, Poff AM, D’Agostino DP, Mukherjee P (2015) Metabolic therapy: a new paradigm for managing malignant brain cancer. Cancer Lett 356:289–300

    Article  CAS  PubMed  Google Scholar 

  91. Johnson BE, Mazor T, Hong C, Barnes M, Aihara K, McLean CY, Fouse SD, Yamamoto S, Ueda H, Tatsuno K, Asthana S, Jalbert LE, Nelson SJ, Bollen AW, Gustafson WC, Charron E, Weiss WA, Smirnov IV, Song JS, Olshen AB, Cha S, Zhao Y, Moore RA, Mungall AJ, Jones SJ, Hirst M, Marra MA, Saito N, Aburatani H, Mukasa A, Berger MS, Chang SM, Taylor BS, Costello JF (2014) Mutational analysis reveals the origin and therapy-driven evolution of recurrent glioma. Science 343:189–193

    Article  CAS  PubMed  Google Scholar 

  92. Klement RJ, Champ CE (2017) Corticosteroids compromise survival in glioblastoma in part through their elevation of blood glucose levels. Brain 140:e16

    PubMed  Google Scholar 

  93. Arcuri C, Tardy M, Rolland B, Armellini R, Menghini AR, Bocchini V (1995) Glutamine synthetase gene expression in a glioblastoma cell-line of clonal origin: regulation by dexamethasone and dibutyryl cyclic AMP. Neurochem Res 20:1133–1139

    Article  CAS  PubMed  Google Scholar 

  94. Wick W, Gorlia T, Bendszus M, Taphoorn M, Sahm F, Harting I, Brandes AA, Taal W, Domont J, Idbaih A, Campone M, Clement PM, Stupp R, Fabbro M, Le Rhun E, Dubois F, Weller M, von Deimling A, Golfinopoulos V, Bromberg JC, Platten M, Klein M, van den Bent MJ (2017) Lomustine and bevacizumab in progressive glioblastoma. N Engl J Med 377:1954–1963

    Article  CAS  PubMed  Google Scholar 

  95. Seyfried TN (2012) Cancer treatment strategies. Cancer as a metabolic disease: on the origin, management, and prevention of cancer. Wiley, Hoboken, NJ, pp 227–289

    Chapter  Google Scholar 

  96. Iwamoto FM, Abrey LE, Beal K, Gutin PH, Rosenblum MK, Reuter VE, DeAngelis LM, Lassman AB (2009) Patterns of relapse and prognosis after bevacizumab failure in recurrent glioblastoma. Neurology 73:1200–1206

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Paez-Ribes M, Allen E, Hudock J, Takeda T, Okuyama H, Vinals F, Inoue M, Bergers G, Hanahan D, Casanovas O (2009) Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer Cell 15:220–231

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. de Groot JF, Fuller G, Kumar AJ, Piao Y, Eterovic K, Ji Y, Conrad CA (2010) Tumor invasion after treatment of glioblastoma with bevacizumab: radiographic and pathologic correlation in humans and mice. Neuro Oncol 12:233–242

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  99. Nanegrungsunk D, Apaijai N, Yarana C, Sripetchwandee J, Limpastan K, Watcharasaksilp W, Vaniyapong T, Chattipakorn N, Chattipakorn SC (2016) Bevacizumab is superior to Temozolomide in causing mitochondrial dysfunction in human brain tumors. Neurol Res 38:285–293

    Article  CAS  PubMed  Google Scholar 

  100. Ratnam NM, Gilbert MR, Giles AJ (2019) Immunotherapy in CNS cancers: the role of immune cell trafficking. Neuro Oncol 21:37–46

    Article  PubMed  Google Scholar 

  101. Ferrara R, Mezquita L, Texier M, Lahmar J, Audigier-Valette C, Tessonnier L, Mazieres J, Zalcman G, Brosseau S, Le Moulec S, Leroy L, Duchemann B, Lefebvre C, Veillon R, Westeel V, Koscielny S, Champiat S, Ferte C, Planchard D, Remon J, Boucher ME, Gazzah A, Adam J, Bria E, Tortora G, Soria JC, Besse B, Caramella C (2018) Hyperprogressive disease in patients with advanced non-small cell lung cancer treated with PD-1/PD-L1 inhibitors or with single-agent chemotherapy. JAMA Oncol 4:1543–1552

    Article  PubMed  PubMed Central  Google Scholar 

  102. Kurz SC, Cabrera LP, Hastie D, Huang R, Unadkat P, Rinne M, Nayak L, Lee EQ, Reardon DA, Wen PY (2018) PD-1 inhibition has only limited clinical benefit in patients with recurrent high-grade glioma. Neurology 91:e1355–e1359

    Article  CAS  PubMed  Google Scholar 

  103. Weller M, Le Rhun E (2019) Immunotherapy for glioblastoma: quo vadis? Nat Rev Clin Oncol

  104. Maroon JC, Seyfried TN, Donohue JP, Bost J (2015) The role of metabolic therapy in treating glioblastoma multiforme. Surg Neurol Int 6:61

    Article  PubMed  PubMed Central  Google Scholar 

  105. Rahbar A, Orrego A, Peredo I, Dzabic M, Wolmer-Solberg N, Straat K, Stragliotto G, Soderberg-Naucler C (2013) Human cytomegalovirus infection levels in glioblastoma multiforme are of prognostic value for survival. J Clin Virol 57:36–42

    Article  PubMed  Google Scholar 

  106. Yu Y, Maguire TG, Alwine JC (2011) Human cytomegalovirus activates glucose transporter 4 expression to increase glucose uptake during infection. J Virol 85:1573–1580

    Article  CAS  PubMed  Google Scholar 

  107. Batich KA, Reap EA, Archer GE, Sanchez-Perez L, Nair SK, Schmittling RJ, Norberg P, Xie W, Herndon JE 2nd, Healy P, McLendon RE, Friedman AH, Friedman HS, Bigner D, Vlahovic G, Mitchell DA, Sampson JH (2017) Long-term survival in glioblastoma with cytomegalovirus pp65-targeted vaccination. Clin Cancer Res 23:1898–1909

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Yu W, Gong JS, Ko M, Garver WS, Yanagisawa K, Michikawa M (2005) Altered cholesterol metabolism in Niemann-Pick type C1 mouse brains affects mitochondrial function. J Biol Chem 280:11731–11739

    Article  CAS  PubMed  Google Scholar 

  109. Yu Y, Clippinger AJ, Alwine JC (2011) Viral effects on metabolism: changes in glucose and glutamine utilization during human cytomegalovirus infection. Trends Microbiol 19:360–367

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Klement RJ, Bandyopadhyay PS, Champ CE, Walach H (2018) Application of Bayesian evidence synthesis to modelling the effect of ketogenic therapy on survival of high grade glioma patients. Theor Biol Med Model 15:12

    Article  PubMed  PubMed Central  Google Scholar 

  111. Noorlag L, De Vos FY, Kok A, Broekman MLD, Seute T, Robe PA, Snijders TJ (2018) Treatment of malignant gliomas with ketogenic or caloric restricted diets: a systematic review of preclinical and early clinical studies. Clin Nutr

  112. Woolf EC, Syed N, Scheck AC (2016) Tumor metabolism, the ketogenic diet and beta-hydroxybutyrate: novel approaches to adjuvant brain tumor therapy. Front Mol Neurosci 9:122

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  113. Varshneya K, Carico C, Ortega A, Patil CG (2015) The efficacy of ketogenic diet and associated hypoglycemia as an adjuvant therapy for high-grade gliomas: a review of the literature. Cureus 7:e251

    PubMed  PubMed Central  Google Scholar 

  114. Strowd RE, Cervenka MC, Henry BJ, Kossoff EH, Hartman AL, Blakeley JO (2015) Glycemic modulation in neuro-oncology: experience and future directions using a modified Atkins diet for high-grade brain tumors. Neurooncol Pract 2:127–136

    PubMed  PubMed Central  Google Scholar 

  115. Rieger J, Steinbach JP (2016) To diet or not to diet—that is still the question. Neuro Oncol 18:1035–1036

    Article  PubMed  PubMed Central  Google Scholar 

  116. Nebeling LC, Miraldi F, Shurin SB, Lerner E (1995) Effects of a ketogenic diet on tumor metabolism and nutritional status in pediatric oncology patients: two case reports. J Am Coll Nutr 14:202–208

    Article  CAS  PubMed  Google Scholar 

  117. Martuscello RT, Vedam-Mai V, McCarthy DJ, Schmoll ME, Jundi MA, Louviere CD, Griffith BG, Skinner CL, Suslov O, Deleyrolle LP, Reynolds BA (2016) A supplemented high-fat low-carbohydrate diet for the treatment of glioblastoma. Clin Cancer Res 22:2482–2495

    Article  CAS  PubMed  Google Scholar 

  118. Winter SF, Loebel F, Dietrich J (2017) Role of ketogenic metabolic therapy in malignant glioma: a systematic review. Crit Rev Oncol Hematol 112:41–58

    Article  PubMed  Google Scholar 

  119. Santos JG, Souza Da Cruz WM, Schonthal AH, Salazar MD, Fontes CA, Qiuirico-Santos T, Da Fonseca CO (2018) Efficacy of a ketogenic diet with concomitant intranasal perillyl alcohol as a novel strategy for the therapy of recurrent glioblastoma. Oncol Lett 15:1263–1270

    PubMed  Google Scholar 

  120. Schwartz KA, Noel M, Nikolai M, Chang HT (2018) Investigating the ketogenic diet as treatment for primary aggressive brain cancer: challenges and lesons learned. Front Nutr 5:1–7

    Article  Google Scholar 

  121. Zhou W, Mukherjee P, Kiebish MA, Markis WT, Mantis JG, Seyfried TN (2007) The calorically restricted ketogenic diet, an effective alternative therapy for malignant brain cancer. Nutr Metab (Lond) 4:5

    Article  CAS  Google Scholar 

  122. Chang HT, Olson LK, Schwartz KA (2013) Ketolytic and glycolytic enzymatic expression profiles in malignant gliomas: implication for ketogenic diet therapy. Nutr Metab 10:47

    Article  CAS  Google Scholar 

  123. Fredericks M, Ramsey RB (1978) 3-Oxo acid coenzyme A transferase activity in brain and tumors of the nervous system. J Neurochem 31:1529–1531

    Article  CAS  PubMed  Google Scholar 

  124. Maurer GD, Brucker DP, Baehr O, Harter PN, Hattingen E, Walenta S, Mueller-Klieser W, Steinbach JP, Rieger J (2011) Differential utilization of ketone bodies by neurons and glioma cell lines: a rationale for ketogenic diet as experimental glioma therapy. BMC Cancer 11:315

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. De Feyter HM, Behar KL, Rao JU, Madden-Hennessey K, Ip KL, Hyder F, Drewes LR, Geschwind JF, de Graaf RA, Rothman DL (2016) A ketogenic diet increases transport and oxidation of ketone bodies in RG2 and 9L gliomas without affecting tumor growth. Neuro Oncol 18:1079–1087

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  126. Schwartz K, Chang HT, Nikolai M, Pernicone J, Rhee S, Olson K, Kurniali PC, Hord NG, Noel M (2015) Treatment of glioma patients with ketogenic diets: report of two cases treated with an IRB-approved energy-restricted ketogenic diet protocol and review of the literature. Cancer Metab 3:3

    Article  PubMed  PubMed Central  Google Scholar 

  127. Szeliga M, Albrecht J (2015) Opposing roles of glutaminase isoforms in determining glioblastoma cell phenotype. Neurochem Int 88:6–9

    Article  CAS  PubMed  Google Scholar 

  128. Nicolay BN, Danielian PS, Kottakis F, Lapek JD Jr, Sanidas I, Miles WO, Dehnad M, Tschop K, Gierut JJ, Manning AL, Morris R, Haigis K, Bardeesy N, Lees JA, Haas W, Dyson NJ (2015) Proteomic analysis of pRb loss highlights a signature of decreased mitochondrial oxidative phosphorylation. Genes Dev 29:1875–1889

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Yang D, Wang MT, Tang Y, Chen Y, Jiang H, Jones TT, Rao K, Brewer GJ, Singh KK, Nie D (2010) Impairment of mitochondrial respiration in mouse fibroblasts by oncogenic H-RAS(Q61L). Cancer Biol Ther 9:122–133

    Article  CAS  PubMed  Google Scholar 

  130. Hu Y, Lu W, Chen G, Wang P, Chen Z, Zhou Y, Ogasawara M, Trachootham D, Feng L, Pelicano H, Chiao PJ, Keating MJ, Garcia-Manero G, Huang P (2012) K-ras(G12V) transformation leads to mitochondrial dysfunction and a metabolic switch from oxidative phosphorylation to glycolysis. Cell Res 22:399–412

    Article  CAS  PubMed  Google Scholar 

  131. Lu W, Pelicano H, Huang P (2010) Cancer metabolism: is glutamine sweeter than glucose? Cancer Cell 18:199–200

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Lu H, Forbes RA, Verma A (2002) Hypoxia-inducible factor 1 activation by aerobic glycolysis implicates the Warburg effect in carcinogenesis. J Biol Chem 277:23111–23115

    Article  CAS  PubMed  Google Scholar 

  133. Yan H, Parsons DW, Jin G, McLendon R, Rasheed BA, Yuan W, Kos I, Batinic-Haberle I, Jones S, Riggins GJ, Friedman H, Friedman A, Reardon D, Herndon J, Kinzler KW, Velculescu VE, Vogelstein B, Bigner DD (2009) IDH1 and IDH2 mutations in gliomas. N Engl J Med 360:765–773

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Mukherjee P, El-Abbadi MM, Kasperzyk JL, Ranes MK, Seyfried TN (2002) Dietary restriction reduces angiogenesis and growth in an orthotopic mouse brain tumour model. Br J Cancer 86:1615–1621

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Mukherjee P, Mulrooney TJ, Marsh J, Blair D, Chiles TC, Seyfried TN (2008) Differential effects of energy stress on AMPK phosphorylation and apoptosis in experimental brain tumor and normal brain. Mol Cancer 7:37

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  136. Mulrooney TJ, Marsh J, Urits I, Seyfried TN, Mukherjee P (2011) Influence of caloric restriction on constitutive expression of NF-kappaB in an experimental mouse astrocytoma. PLoS ONE 6:e18085

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Shelton LM, Huysentruyt LC, Mukherjee P, Seyfried TN (2010) Calorie restriction as an anti-invasive therapy for malignant brain cancer in the VM mouse. ASN Neuro 2:e00038

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  138. Maalouf M, Sullivan PG, Davis L, Kim DY, Rho JM (2007) Ketones inhibit mitochondrial production of reactive oxygen species production following glutamate excitotoxicity by increasing NADH oxidation. Neuroscience 145:256–264

    Article  CAS  PubMed  Google Scholar 

  139. Veech RL (2004) The therapeutic implications of ketone bodies: the effects of ketone bodies in pathological conditions: ketosis, ketogenic diet, redox states, insulin resistance, and mitochondrial metabolism. Prostaglandins Leukot Essent Fatty Acids 70:309–319

    Article  CAS  PubMed  Google Scholar 

  140. D’Agostino DP, Olson JE, Dean JB (2009) Acute hyperoxia increases lipid peroxidation and induces plasma membrane blebbing in human U87 glioblastoma cells. Neuroscience 159:1011–1022

    Article  PubMed  CAS  Google Scholar 

  141. Veech RL, Todd King M, Pawlosky R, Kashiwaya Y, Bradshaw PC, Curtis W (2019) The “great” controlling nucleotide coenzymes. IUBMB Life

  142. Rieger J, Bahr O, Maurer GD, Hattingen E, Franz K, Brucker D, Walenta S, Kammerer U, Coy JF, Weller M, Steinbach JP (2014) ERGO: a pilot study of ketogenic diet in recurrent glioblastoma. Int J Oncol 44:1843–1852

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Champ CE, Palmer JD, Volek JS, Werner-Wasik M, Andrews DW, Evans JJ, Glass J, Kim L, Shi W (2014) Targeting metabolism with a ketogenic diet during the treatment of glioblastoma multiforme. J Neurooncol 117:125–131

    Article  CAS  PubMed  Google Scholar 

  144. Meidenbauer JJ, Mukherjee P, Seyfried TN (2015) The glucose ketone index calculator: a simple tool to monitor therapeutic efficacy for metabolic management of brain cancer. Nutr Metab (Lond) 12:12

    Article  CAS  Google Scholar 

  145. Klement RJ (2017) Beneficial effects of ketogenic diets for cancer patients: a realist review with focus on evidence and confirmation. Med Oncol 34:132

    Article  PubMed  Google Scholar 

  146. Iyikesici MS, Slocum AK, Slocum A, Berkarda FB, Kalamian M, Seyfried TN (2017) Efficacy of metabolically supported chemotherapy combined with ketogenic diet, hyperthermia, and hyperbaric oxygen therapy for stage IV triple-negative breast cancer. Cureus 9:e1445

    PubMed  PubMed Central  Google Scholar 

  147. Poff AM, Ari C, Seyfried TN, D’Agostino DP (2013) The ketogenic diet and hyperbaric oxygen therapy prolong survival in mice with systemic metastatic cancer. PLoS ONE 8:e65522

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Husain Z, Huang Y, Seth P, Sukhatme VP (2013) Tumor-derived lactate modifies antitumor immune response: effect on myeloid-derived suppressor cells and NK cells. Journal of immunology 191:1486–1495

    Article  CAS  Google Scholar 

  149. Denny CA, Heinecke KA, Kim YP, Baek RC, Loh KS, Butters TD, Bronson RT, Platt FM, Seyfried TN (2010) Restricted ketogenic diet enhances the therapeutic action of N-butyldeoxynojirimycin towards brain GM2 accumulation in adult Sandhoff disease mice. J Neurochem 113:1525–1535

    CAS  PubMed  Google Scholar 

  150. Ye H, Chen M, Cao F, Huang H, Zhan R, Zheng X (2016) Chloroquine, an autophagy inhibitor, potentiates the radiosensitivity of glioma initiating cells by inhibiting autophagy and activating apoptosis. BMC Neurol 16:178

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  151. Al-Bari MA (2015) Chloroquine analogues in drug discovery: new directions of uses, mechanisms of actions and toxic manifestations from malaria to multifarious diseases. J Antimicrob Chemother 70:1608–1621

    PubMed  Google Scholar 

  152. Hrabak A, Sefrioui H, Vercruysse V, Temesi A, Bajor T, Vray B (1998) Action of chloroquine on nitric oxide production and parasite killing by macrophages. Eur J Pharmacol 354:83–90

    Article  CAS  PubMed  Google Scholar 

  153. Yang C, Ko B, Hensley CT, Jiang L, Wasti AT, Kim J, Sudderth J, Calvaruso MA, Lumata L, Mitsche M, Rutter J, Merritt ME, DeBerardinis RJ (2014) Glutamine oxidation maintains the TCA cycle and cell survival during impaired mitochondrial pyruvate transport. Mol Cell 56:414–424

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Zuccoli G, Marcello N, Pisanello A, Servadei F, Vaccaro S, Mukherjee P, Seyfried TN (2010) Metabolic management of glioblastoma multiforme using standard therapy together with a restricted ketogenic diet: case report. Nutr Metab (Lond) 7:33

    Article  CAS  Google Scholar 

  155. Kalamian M (2017) KETO for CANCER: ketogenic metabolic therapy as a targeted nutritional strategy. Chelsea Green, White River Junction, VT

    Google Scholar 

  156. Winter SF, Loebel F, Loeffler J, Batchelor TT, Martinez-Lage M, Vajkoczy P, Dietrich J (2019) Treatment-induced brain tissue necrosis: a clinical challenge in neuro-oncology. Neuro Oncol

  157. Warburg O (1956) On the respiratory impairment in cancer cells. Science 124:269–270

    CAS  PubMed  Google Scholar 

  158. Seyfried TN, Sanderson TM, El-Abbadi MM, McGowan R, Mukherjee P (2003) Role of glucose and ketone bodies in the metabolic control of experimental brain cancer. Br J Cancer 89:1375–1382

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Meidenbauer JJ, Ta N, Seyfried TN (2014) Influence of a ketogenic diet, fish-oil, and calorie restriction on plasma metabolites and lipids in C57BL/6J mice. Nutr Metab 11:23

    Article  CAS  Google Scholar 

  160. Kiebish MA, Han X, Cheng H, Lunceford A, Clarke CF, Moon H, Chuang JH, Seyfried TN (2008) Lipidomic analysis and electron transport chain activities in C57BL/6J mouse brain mitochondria. J Neurochem 106:299–312

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Kiebish MA, Han X, Cheng H, Seyfried TN (2009) In vitro growth environment produces lipidomic and electron transport chain abnormalities in mitochondria from non-tumorigenic astrocytes and brain tumours. ASN Neuro 1:e00011

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  162. Wise DR, DeBerardinis RJ, Mancuso A, Sayed N, Zhang XY, Pfeiffer HK, Nissim I, Daikhin E, Yudkoff M, McMahon SB, Thompson CB (2008) Myc regulates a transcriptional program that stimulates mitochondrial glutaminolysis and leads to glutamine addiction. Proc Natl Acad Sci USA 105:18782–18787

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Yang I, Aghi MK (2009) New advances that enable identification of glioblastoma recurrence. Nat Rev Clin Oncol 6:648–657

    Article  PubMed  Google Scholar 

  164. Spence AM, Muzi M, Graham MM, O’Sullivan F, Krohn KA, Link JM, Lewellen TK, Lewellen B, Freeman SD, Berger MS, Ojemann GA (1998) Glucose metabolism in human malignant gliomas measured quantitatively with PET, 1-[C-11]glucose and FDG: analysis of the FDG lumped constant. J Nucl Med 39:440–448

    CAS  PubMed  Google Scholar 

  165. Seyfried TN, Mukherjee P (2005) Targeting energy metabolism in brain cancer: review and hypothesis. Nutr Metab (Lond) 2:30

    Article  CAS  Google Scholar 

  166. McKenna MC, Gruetter R, Sonnewald U, Waagepetersen HS, Schousboe A (2006) Energy Metabolism of the Brain. In: Siegel GJ, Albers RW, Bradey ST, Price DP (eds) Basic neurochemistry: molecular, cellular, and medical aspects. Elsevier Academic Press, New York, pp 531–557

    Google Scholar 

  167. Sonnewald U, Schousboe A (2016) Introduction to the glutamate-glutamine cycle. Adv Neurobiol 13:1–7

    Article  PubMed  Google Scholar 

  168. Newsholme P (2001) Why is l-glutamine metabolism important to cells of the immune system in health, postinjury, surgery or infection? J Nutr 131:2515S–2522S discussion 2523S–2514S

    Article  CAS  PubMed  Google Scholar 

  169. Lewis C, Murdoch C (2005) Macrophage responses to hypoxia: implications for tumor progression and anti-cancer therapies. Am J Pathol 167:627–635

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Dix AR, Brooks WH, Roszman TL, Morford LA (1999) Immune defects observed in patients with primary malignant brain tumors. J Neuroimmunol 100:216–232

    Article  CAS  PubMed  Google Scholar 

  171. Seyfried TN, Kiebish MA, Marsh J, Shelton LM, Huysentruyt LC, Mukherjee P (2011) Metabolic management of brain cancer. Biochem Biophys Acta 1807:577–594

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The author would like to acknowledge support from the Foundation for Metabolic Cancer Therapies, the Claudia & Nelson Pleltz Foundation, Crossfit Inc., Lewis Topper, Edward Miller, Ellen Davis, and the Boston College research expense fund. The authors also thank Pedro Arteaga, Norkys Sanchez, and Gianny Arteaga (Maracaibo-Venezuela) for technical help with EM micrographs.

Funding

Funding was provided by Foundation for Metabolic Cancer Therapies (Grant No. 5101551), Claudia & Nelson Peltz Foundation and CrossFit (Grant No. 51o5681).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Thomas N. Seyfried.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Special issue in honour of Professor Vera Adam-Vizi.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Seyfried, T.N., Shelton, L., Arismendi-Morillo, G. et al. Provocative Question: Should Ketogenic Metabolic Therapy Become the Standard of Care for Glioblastoma?. Neurochem Res 44, 2392–2404 (2019). https://doi.org/10.1007/s11064-019-02795-4

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11064-019-02795-4

Keywords

Navigation