The ketogenic diet is an effective adjuvant to radiation therapy for the treatment of malignant glioma

Mohammed G Abdelwahab, Kathryn E Fenton, Mark C Preul, Jong M Rho, Andrew Lynch, Phillip Stafford, Adrienne C Scheck, Mohammed G Abdelwahab, Kathryn E Fenton, Mark C Preul, Jong M Rho, Andrew Lynch, Phillip Stafford, Adrienne C Scheck

Abstract

Introduction: The ketogenic diet (KD) is a high-fat, low-carbohydrate diet that alters metabolism by increasing the level of ketone bodies in the blood. KetoCal® (KC) is a nutritionally complete, commercially available 4:1 (fat:carbohydrate+protein) ketogenic formula that is an effective non-pharmacologic treatment for the management of refractory pediatric epilepsy. Diet-induced ketosis causes changes to brain homeostasis that have potential for the treatment of other neurological diseases such as malignant gliomas.

Methods: We used an intracranial bioluminescent mouse model of malignant glioma. Following implantation animals were maintained on standard diet (SD) or KC. The mice received 2×4 Gy of whole brain radiation and tumor growth was followed by in vivo imaging.

Results: Animals fed KC had elevated levels of β-hydroxybutyrate (p = 0.0173) and an increased median survival of approximately 5 days relative to animals maintained on SD. KC plus radiation treatment were more than additive, and in 9 of 11 irradiated animals maintained on KC the bioluminescent signal from the tumor cells diminished below the level of detection (p<0.0001). Animals were switched to SD 101 days after implantation and no signs of tumor recurrence were seen for over 200 days.

Conclusions: KC significantly enhances the anti-tumor effect of radiation. This suggests that cellular metabolic alterations induced through KC may be useful as an adjuvant to the current standard of care for the treatment of human malignant gliomas.

Conflict of interest statement

Competing Interests: This work was supported by a grant from Nutricia Advanced Medical Nutrition through Nutricia North America, the company that distributes KetoCal® in the USA. Dr. Andrew Lynch is employed by Nutricia Advanced Medical Nutrition in the UK. Dr. Lynch did provide input to the study design. This does not alter the authors' adherence to all the PLoS ONE policies on sharing data and materials.

Figures

Figure 1. KetoCal® enhances survival of glioma-bearing…
Figure 1. KetoCal® enhances survival of glioma-bearing mice.
Kaplan-Meier plot of survival in KC versus SD (A), radiation versus KC plus radiation (B). Animals on KC survived significantly longer when treated with KC alone (p

Figure 2. KetoCal® plus radiation treated animals…

Figure 2. KetoCal® plus radiation treated animals experienced a loss of bioluminescence.

(A) Radiation and…

Figure 2. KetoCal® plus radiation treated animals experienced a loss of bioluminescence.
(A) Radiation and KetoCal® plus radiation animal imaged every three days. Color scheme represents signal intensity with red representing the highest intensity photon count. (B) (C) Bioluminescent signal plotted as in vivo photon count versus days post-implantation.

Figure 3. Blood ketone and glucose levels.

Figure 3. Blood ketone and glucose levels.

Measurements of animal ketone and glucose levels show…

Figure 3. Blood ketone and glucose levels.
Measurements of animal ketone and glucose levels show higher βHB blood levels in animals treated with KetoCal®. (A) βHB levels (B) Blood glucose measurements. ** = p

Figure 4. Animal weights.

Weight measurements were…

Figure 4. Animal weights.

Weight measurements were taken every 3 days. Graph shows animals weights…

Figure 4. Animal weights.
Weight measurements were taken every 3 days. Graph shows animals weights normalized to the average starting weight of each group on day zero. (B) Arrow denotes a long-term survivor 60 days following tumor implantation in the SD plus radiation group.
Similar articles
Cited by
References
    1. Ruggiero A, Cefalo G, Garre ML, Massimino M, Colosimo C, et al. Phase II trial of temozolomide in children with recurrent high-grade glioma. J Neurooncol. 2006;77:89–94. - PubMed
    1. Nicholas MK, Lukas RV, Chmura S, Yamini B, Lesniak M, et al. Molecular heterogeneity in glioblastoma: therapeutic opportunities and challenges. Semin Oncol. 2011;38:243–253. - PubMed
    1. Niyazi M, Siefert A, Schwarz SB, Ganswindt U, Kreth FW, et al. Therapeutic options for recurrent malignant glioma. Radiother Oncol. 2011;98:1–14. - PubMed
    1. Roesler R, Brunetto AT, Abujamra AL, de Farias CB, Brunetto AL, et al. Current and emerging molecular targets in glioma. Expert Rev Anticancer Ther. 2010;10:1735–1751. - PubMed
    1. Warburg O. On the origin of cancer cells. Science. 1956;123:309–314. - PubMed
Show all 38 references
Publication types
MeSH terms
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 2. KetoCal® plus radiation treated animals…
Figure 2. KetoCal® plus radiation treated animals experienced a loss of bioluminescence.
(A) Radiation and KetoCal® plus radiation animal imaged every three days. Color scheme represents signal intensity with red representing the highest intensity photon count. (B) (C) Bioluminescent signal plotted as in vivo photon count versus days post-implantation.
Figure 3. Blood ketone and glucose levels.
Figure 3. Blood ketone and glucose levels.
Measurements of animal ketone and glucose levels show higher βHB blood levels in animals treated with KetoCal®. (A) βHB levels (B) Blood glucose measurements. ** = p

Figure 4. Animal weights.

Weight measurements were…

Figure 4. Animal weights.

Weight measurements were taken every 3 days. Graph shows animals weights…

Figure 4. Animal weights.
Weight measurements were taken every 3 days. Graph shows animals weights normalized to the average starting weight of each group on day zero. (B) Arrow denotes a long-term survivor 60 days following tumor implantation in the SD plus radiation group.
Figure 4. Animal weights.
Figure 4. Animal weights.
Weight measurements were taken every 3 days. Graph shows animals weights normalized to the average starting weight of each group on day zero. (B) Arrow denotes a long-term survivor 60 days following tumor implantation in the SD plus radiation group.

References

    1. Ruggiero A, Cefalo G, Garre ML, Massimino M, Colosimo C, et al. Phase II trial of temozolomide in children with recurrent high-grade glioma. J Neurooncol. 2006;77:89–94.
    1. Nicholas MK, Lukas RV, Chmura S, Yamini B, Lesniak M, et al. Molecular heterogeneity in glioblastoma: therapeutic opportunities and challenges. Semin Oncol. 2011;38:243–253.
    1. Niyazi M, Siefert A, Schwarz SB, Ganswindt U, Kreth FW, et al. Therapeutic options for recurrent malignant glioma. Radiother Oncol. 2011;98:1–14.
    1. Roesler R, Brunetto AT, Abujamra AL, de Farias CB, Brunetto AL, et al. Current and emerging molecular targets in glioma. Expert Rev Anticancer Ther. 2010;10:1735–1751.
    1. Warburg O. On the origin of cancer cells. Science. 1956;123:309–314.
    1. Liang BC, Grootveld M. The importance of mitochondria in the tumourigenic phenotype: gliomas as the paradigm (review). Int J Mol Med. 2011;27:159–171.
    1. Robey RB, Hay N. Is Akt the “Warburg kinase”?-Akt-energy metabolism interactions and oncogenesis. Semin Cancer Biol. 2009;19:25–31.
    1. Lund TM, Risa O, Sonnewald U, Schousboe A, Waagepetersen HS. Availability of neurotransmitter glutamate is diminished when beta-hydroxybutyrate replaces glucose in cultured neurons. J Neurochem. 2009;110:80–91.
    1. Wallace DC, Fan W, Procaccio V. Mitochondrial energetics and therapeutics. Annu Rev Pathol. 2010;5:297–348:297–348.
    1. Samala R, Klein J, Borges K. The ketogenic diet changes metabolite levels in hippocampal extracellular fluid. Neurochem Int. 2011;58:5–8.
    1. Schiff M, Benit P, Coulibaly A, Loublier S, El-Khoury R, et al. Mitochondrial response to controlled nutrition in health and disease. Nutr Rev. 2011;69:65–75.
    1. Eloqayli H, Melo TM, Haukvik A, Sonnewald U. [2,4-(13)C]beta-hydroxybutyrate metabolism in astrocytes and C6 glioblastoma cells. Neurochem Res. 2011;36:1566–1573.
    1. Stafford P, Abdelwahab MG, Kim DY, Preul MC, Rho JM, et al. The ketogenic diet reverses gene expression patterns and reduces reactive oxygen species levels when used as an adjuvant therapy for glioma. Nutr Metab (Lond) 2010;7:74.
    1. Mukherjee P, Abate LE, Seyfried TN. Antiangiogenic and proapoptotic effects of dietary restriction on experimental mouse and human brain tumors. Clin Cancer Res. 2004;10:5622–5629.
    1. Mukherjee P, El-Abbadi MM, Kasperzyk JL, Ranes MK, Seyfried TN. Dietary restriction reduces angiogenesis and growth in an orthotopic mouse brain tumour model. Br J Cancer. 2002;86:1615–1621.
    1. Mulrooney TJ, Marsh J, Urits I, Seyfried TN, Mukherjee P. Influence of caloric restriction on constitutive expression of NF-kappaB in an experimental mouse astrocytoma. PLoS ONE. 2011;6:e18085.
    1. Baranano KW, Hartman AL. The ketogenic diet: uses in epilepsy and other neurologic illnesses. Curr Treat Options Neurol. 2008;10:410–419.
    1. Zuccoli G, Marcello N, Pisanello A, Servadei F, Vaccaro S, et al. Metabolic management of glioblastoma multiforme using standard therapy together with a restricted ketogenic diet: Case Report. Nutrition and Metabolism. 2010;7:33–53.
    1. Zhou W, Mukherjee P, Kiebish MA, Markis WT, Mantis JG, et al. The calorically restricted ketogenic diet, an effective alternative therapy for malignant brain cancer. Nutr Metab (Lond) 2007;4:5:5.
    1. Seyfried TN, Sanderson TM, El-Abbadi MM, McGowan R, Mukherjee P. Role of glucose and ketone bodies in the metabolic control of experimental brain cancer. Br J Cancer. 2003;89:1375–1382.
    1. Seyfried TN, Mukherjee P. Targeting energy metabolism in brain cancer: review and hypothesis. Nutrition and Metabolism. 2005;2:30–38.
    1. Maurer GD, Brucker DP, Bahr O, Harter PN, Hattingen E, 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;11:315:315.
    1. Skinner R, Trujillo A, Ma X, Beierle EA. Ketone bodies inhibit the viability of human neuroblastoma cells. J Pediatr Surg. 2009;44:212–216.
    1. Bonuccelli G, Tsirigos A, Whitaker-Menezes D, Pavlides S, Pestell RG, et al. Ketones and lactate “fuel” tumor growth and metastasis: Evidence that epithelial cancer cells use oxidative mitochondrial metabolism. Cell Cycle. 2010;9:3506–3514.
    1. Seyfried TN, Shelton LM, Mukherjee P. Does the existing standard of care increase glioblastoma energy metabolism? Lancet Oncol. 2010;11:811–813.
    1. Yamada M, Tomida A, Yun J, Cai B, Yoshikawa H, et al. Cellular sensitization to cisplatin and carboplatin with decreased removal of platinum-DNA adduct by glucose-regulated stress. Cancer Chemother Pharmacol. 1999;44:59–64.
    1. Dwarakanath B, Jain V. Targeting glucose metabolism with 2-deoxy-D-glucose for improving cancer therapy. Future Oncol. 2009;5:581–585.
    1. Marsh J, Mukherjee P, Seyfried TN. Drug/diet synergy for managing malignant astrocytoma in mice: 2-deoxy-D-glucose and the restricted ketogenic diet. Nutr Metab (Lond) 2008;5:33:33.
    1. Maschek G, Savaraj N, Priebe W, Braunschweiger P, Hamilton K, 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;64:31–34.
    1. Ralser M, Wamelink MM, Struys EA, Joppich C, Krobitsch S, et al. A catabolic block does not sufficiently explain how 2-deoxy-D-glucose inhibits cell growth. Proc Natl Acad Sci U S A. 2008;105:17807–17811.
    1. Gupta S, Farooque A, Adhikari JS, Singh S, Dwarakanath BS. Enhancement of radiation and chemotherapeutic drug responses by 2-deoxy-D-glucose in animal tumors. J Cancer Res Ther. 2009;5(Suppl 1):S16–S20.
    1. Chautard E, Loubeau G, Tchirkov A, Chassagne J, Vermot-Desroches C, et al. Akt signaling pathway: a target for radiosensitizing human malignant glioma. Neuro Oncol. 2010;12:434–443.
    1. Li HF, Kim JS, Waldman T. Radiation-induced Akt activation modulates radioresistance in human glioblastoma cells. Radiat Oncol. 2009;4:43.
    1. Nebeling LC, Lerner E. Implementing a ketogenic diet based on medium-chain triglyceride oil in pediatric patients with cancer. J Am Diet Assoc. 1995;95:693–697.
    1. Nebeling LC, Miraldi F, Shurin SB, Lerner E. Effects of a ketogenic diet on tumor metabolism and nutritional status in pediatric oncology patients: two case reports. J Am Coll Nutr. 1995;14:202–208.
    1. Abdelwahab MG, Sankar T, Preul MC, Scheck AC. Intracranial Implantation with Subsequent In Vivo Bioluminescent Imaging of Murine Gliomas. JOVE. 2011;57:e3403.
    1. Newcomb EW, Demaria S, Lukyanov Y, Shao Y, Schnee T, et al. The combination of ionizing radiation and peripheral vaccination produces long-term survival of mice bearing established invasive GL261 gliomas. Clin Cancer Res. 2006;12:4730–4737.
    1. Jouanneau E, Poujol D, Gulia S, Le MI, Blay JY, et al. Dendritic cells are essential for priming but inefficient for boosting antitumour immune response in an orthotopic murine glioma model. Cancer Immunol Immunother. 2006;55:254–267.

Source: PubMed

3
구독하다