ERGO: a pilot study of ketogenic diet in recurrent glioblastoma

Johannes Rieger, Oliver Bähr, Gabriele D Maurer, Elke Hattingen, Kea Franz, Daniel Brucker, Stefan Walenta, Ulrike Kämmerer, Johannes F Coy, Michael Weller, Joachim P Steinbach, Johannes Rieger, Oliver Bähr, Gabriele D Maurer, Elke Hattingen, Kea Franz, Daniel Brucker, Stefan Walenta, Ulrike Kämmerer, Johannes F Coy, Michael Weller, Joachim P Steinbach

Abstract

Limiting dietary carbohydrates inhibits glioma growth in preclinical models. Therefore, the ERGO trial (NCT00575146) examined feasibility of a ketogenic diet in 20 patients with recurrent glioblastoma. Patients were put on a low-carbohydrate, ketogenic diet containing plant oils. Feasibility was the primary endpoint, secondary endpoints included the percentage of patients reaching urinary ketosis, progression-free survival (PFS) and overall survival. The effects of a ketogenic diet alone or in combination with bevacizumab was also explored in an orthotopic U87MG glioblastoma model in nude mice. Three patients (15%) discontinued the diet for poor tolerability. No serious adverse events attributed to the diet were observed. Urine ketosis was achieved at least once in 12 of 13 (92%) evaluable patients. One patient achieved a minor response and two patients had stable disease after 6 weeks. Median PFS of all patients was 5 (range, 3-13) weeks, median survival from enrollment was 32 weeks. The trial allowed to continue the diet beyond progression. Six of 7 (86%) patients treated with bevacizumab and diet experienced an objective response, and median PFS on bevacizumab was 20.1 (range, 12-124) weeks, for a PFS at 6 months of 43%. In the mouse glioma model, ketogenic diet alone had no effect on median survival, but increased that of bevacizumab-treated mice from 52 to 58 days (p<0.05). In conclusion, a ketogenic diet is feasible and safe but probably has no significant clinical activity when used as single agent in recurrent glioma. Further clinical trials are necessary to clarify whether calorie restriction or the combination with other therapeutic modalities, such as radiotherapy or anti-angiogenic treatments, could enhance the efficacy of the ketogenic diet.

Figures

Figure 1.
Figure 1.
Study design and course of treatment. (A) After study inclusion, patients were treated with the ketogenic diet. At 6 weeks on the diet or in the presence of clinical signs of deterioration, MRI was performed. In case of at least stable disease, diet was continued. In case of a progression, the patients were offered salvage therapy while continuing the diet. (B) Flowchart diagram showing the course of dietary treatment in the 20 included patients.
Figure 2.
Figure 2.
Urine ketosis. The fraction of ketone body-positive urine analyses (ketone bodies >0.5 mmol/l) is reported in the 13 patients with these tests available. Each bar represents the value of one patient, sorted from lowest to the highest ratios (left to right). The individual values are also presented above each bar.
Figure 3.
Figure 3.
Patients’ self reported rating of diet tolerability. The patients were asked to fill out a self-reported questionnaire for possible diet-related side-effects once every week on diet. These sheets were completed at least once in 12 patients. Shown are the ratings of each patient in the categories diarrhea (A), constipation (B), hunger (C) and demand for glucose (D) in every week (dot) and the mean value of these ratings in the corresponding week (line). The scale was defined as follows: 0, not present; 1, weak; 2, moderate; 3, strong. Questionnaires were available in 7–11 patients at the shown time-points.
Figure 4.
Figure 4.
MRI finding in 3 patients. (A) T1-weighted images after intravenous application of gadolinium-DTPA (Gd-DTPA) before (left panel) and after 6 weeks on the ketogenic diet (KD) (right panel) in axial (upper) and coronar (lower) planes of the patient with a minor response. (B) T1-weighted images after intravenous application of Gd-DTPA before (left panel) and after 6 weeks on the ketogenic diet (right panel) in axial directions of a patient with stable disease. (C) Fluid-atenuated inversion recovery (FLAIR) images and T1-weighted images after application of Gd-DTPA before (left panel) and 6 weeks after start of bevacizumab (BEV) in a patient with partial response to bevacizumab who continued the diet.
Figure 5.
Figure 5.
Analysis of PFS. (A) PFS of patients on diet who achieved stable ketosis as defined as urine ketosis in >50% of measurements (n=8) compared to those patients who did not achieve stabile ketosis (n=5) was analysed by Kaplan-Meier analysis (p=0.069, log-rank-test). (B) PFS of patients who received bevacizumab while on ketogenic diet (n=7) vs. a cohort of patients who were treated in the same period of time with bevacizumab but without ketogenic diet (n=28) (p=0.38, log-rank test).
Figure 6.
Figure 6.
Mouse xenograft experiments. (A) 3-OHB levels in the blood were determined 24 days after U87MG tumor cell inoculation in mice on standard diet (SD) or ketogenic diet (KD) treated with PBS (control) or bevacizumab (BEV). (B) Kaplan-Meier analysis of the treatment groups. (C) At day 28 after tumor cell inoculation, 3 mice per group were analysed by MRI. Upper panel, representative T2-weighted images and T1-weighted images after infusion of Gd-DTPA are shown. Lower panel, tumor volumes were measured as described, and individual (dot) and mean values (bar) are indicated. (D) Upper panel, representative hematoxylin-eosin stainings (H&E) and color-coded distribution of ATP concentrations in representative slices. Lower panel, ATP content in the tumor areas was quantified, and individual (dot) and mean values (bar) are shown.

References

    1. Warburg O, Posener K, Negelein E. Ueber den Stoffwechsel der Tumoren. Biochem Z. 1924;152:319–344. (in German)
    1. TCGA. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature. 2008;455:1061–1068.
    1. Elstrom RL, Bauer DE, Buzzai M, Karnauskas R, Harris MH, Plas DR, Zhuang H, Cinalli RM, Alavi A, Rudin CM, Thompson CB. Akt stimulates aerobic glycolysis in cancer cells. Cancer Res. 2004;64:3892–3899.
    1. Matoba S, Kang J, Patino WD, Wragg A, Boehm M, Gavrilova O, Hurley PJ, Bunz F, Hwang PM. p53 regulates mitochondrial respiration. Science. 2006;312:1650–1653.
    1. Wanka C, Brucker DP, Bähr O, Ronellenfitsch M, Weller M, Steinbach JP, Rieger J. Synthesis of cytochrome c oxidase 2: a p53-dependent metabolic regulator that promotes respiratory function and protects glioma and colon cancer cells from hypoxia-induced cell death. Oncogene. 2012;31:3764–3776.
    1. Bensaad K, Tsuruta A, Selak MA, Vidal MNC, Nakano K, Bartrons R, Gottlieb E, Vousden KH. TIGAR, a p53-inducible regulator of glycolysis and apoptosis. Cell. 2006;126:107–120.
    1. Wanka C, Steinbach JP, Rieger J. Tp53-induced glycolysis and apoptosis regulator (TIGAR) protects glioma cells from starvation-induced cell death by upregulating respiration and improving cellular redox homeostasis. J Biol Chem. 2012;287:33436–33446.
    1. Denko NC. Hypoxia, HIF1 and glucose metabolism in the solid tumour. Nat Rev Cancer. 2008;8:705–713.
    1. Papandreou I, Cairns RA, Fontana L, Lim AL, Denko NC. HIF-1 mediates adaptation to hypoxia by actively down-regulating mitochondrial oxygen consumption. Cell Metab. 2006;3:187–197.
    1. Liu Y, Li Y, Tian R, Liu W, Fei Z, Long Q, Wang X, Zhang X. The expression and significance of HIF-1alpha and GLUT-3 in glioma. Brain Res. 2009;1304:149–154.
    1. Roslin M, Henriksson R, Bergström P, Ungerstedt U, Bergenheim AT. Baseline levels of glucose metabolites, glutamate and glycerol in malignant glioma assessed by stereo-tactic microdialysis. J Neurooncol. 2003;61:151–160.
    1. Padma MV, Said S, Jacobs M, Hwang DR, Dunigan K, Satter M, Christian B, Ruppert J, Bernstein T, Kraus G, Mantil JC. Prediction of pathology and survival by FDG PET in gliomas. J Neurooncol. 2003;64:227–237.
    1. Hirata K, Terasaka S, Shiga T, Hattori N, Magota K, Kobayashi H, Yamaguchi S, Houkin K, Tanaka S, Kuge Y, Tamaki N. (18)F-Fluoromisonidazole positron emission tomography may differentiate glioblastoma multiforme from less malignant gliomas. Eur J Nucl Med Mol Imaging. 2012;39:760–770.
    1. Maher EA, Marin-Valencia I, Bachoo RM, Mashimo T, Raisanen J, Hatanpaa KJ, Jindal A, Jeffrey FM, Choi C, Madden C, Mathews D, Pascual JM, Mickey BE, Malloy CR, Deberardinis RJ. Metabolism of [U-(13) C]glucose in human brain tumors in vivo. NMR Biomed. 2012;25:1234–1244.
    1. Marin-Valencia I, Yang C, Mashimo T, Cho S, Baek H, Yang X, Rajagopalan KN, Maddie M, Vemireddy V, Zhao Z, Cai L, Good L, Tu BP, Hatanpaa KJ, Mickey BE, Matés JM, Pascual JM, Maher EA, Malloy CR, Deberardinis RJ, Bachoo RM. Analysis of tumor metabolism reveals mitochondrial glucose oxidation in genetically diverse human glioblastomas in the mouse brain in vivo. Cell Metab. 2012;15:827–837.
    1. Ward PS, Thompson CB. Metabolic reprogramming: a cancer hallmark even Warburg did not anticipate. Cancer Cell. 2012;21:297–308.
    1. Jeon S, Chandel NS, Hay N. AMPK regulates NADPH homeostasis to promote tumour cell survival during energy stress. Nature. 2012;485:661–665.
    1. Goodwin PJ, Ennis M, Pritchard KI, Trudeau ME, Koo J, Madarnas Y, Hartwick W, Hoffman B, Hood N. Fasting insulin and outcome in early-stage breast cancer: results of a prospective cohort study. J Clin Oncol. 2002;20:42–51.
    1. Wolk A, Mantzoros CS, Andersson SO, Bergström R, Signorello LB, Lagiou P, Adami HO, Trichopoulos D. Insulin-like growth factor 1 and prostate cancer risk: a population-based, case-control study. J Natl Cancer Inst. 1998;90:911–915.
    1. Allen NE, Key TJ, Appleby PN, Travis RC, Roddam AW, Rinaldi S, Egevad L, Rohrmann S, Linseisen J, Pischon T, Boeing H, Johnsen NF, Tjønneland A, Grønbaek H, Overvad K, Kiemeney L, Bueno-de-Mesquita HB, Bingham S, Khaw KT, Tumino R, Berrino F, Mattiello A, Sacerdote C, Palli D, Quirós JR, Ardanaz E, Navarro C, Larrañaga N, Gonzalez C, Sanchez M, Trichopoulou A, Travezea C, Trichopoulos D, Jenab M, Ferrari P, Riboli E, Kaaks R. Serum insulin-like growth factor (IGF)-I and IGF-binding protein-3 concentrations and prostate cancer risk: results from the European Prospective Investigation into Cancer and Nutrition. Cancer Epidemiol Biomarkers Prev. 2007;16:1121–1127.
    1. Renehan AG, Egger M, Minder C, O’Dwyer ST, Shalet SM, Zwahlen M. IGF-I, IGF binding protein-3 and breast cancer risk: comparison of 3 meta-analyses. Int J Cancer. 115:1006–1007. author reply, 1008, 2005.
    1. Bowker SL, Majumdar SR, Veugelers P, Johnson JA. Increased cancer-related mortality for patients with type 2 diabetes who use sulfonylureas or insulin: response to Farooki and Schneider. Diabetes Care. 2006;29:1990–1991.
    1. Derr RL, Ye X, Islas MU, Desideri S, Saudek CD, Grossman SA. Association between hyperglycemia and survival in patients with newly diagnosed glioblastoma. J Clin Oncol. 2009;27:1082–1086.
    1. Zhou W, Mukherjee P, Kiebish MA, Markis WT, Mantis JG, Seyfried TN. The calorically restricted ketogenic diet, an effective alternative therapy for malignant brain cancer. Nutr Metab (Lond) 2007;4:5.
    1. Marsh J, Mukherjee P, Seyfried TN. Akt-dependent proapoptotic effects of dietary restriction on late-stage management of a phosphatase and tensin homologue/tuberous sclerosis complex 2-deficient mouse astrocytoma. Clin Cancer Res. 2008;14:7751–7762.
    1. Stafford P, Abdelwahab MG, Kim DY, Preul MC, Rho JM, Scheck AC. The ketogenic diet reverses gene expression patterns and reduces reactive oxygen species levels when used as an adjuvant therapy for glioma. Nutr Metab (Lond) 774:2010.
    1. Kossoff EH, Rowley H, Sinha SR, Vining EPG. A prospective study of the modified Atkins diet for intractable epilepsy in adults. Epilepsia. 2008;49:316–319.
    1. Shai I, Schwarzfuchs D, Henkin Y, Shahar DR, Witkow S, Greenberg I, Golan R, Fraser D, Bolotin A, Vardi H, Tangi-Rozental O, Zuk-Ramot R, Sarusi B, Brickner D, Schwartz Z, Sheiner E, Marko R, Katorza E, Thiery J, Fiedler GM, Blüher M, Stumvoll M, Stampfer MJ. Weight loss with a low-carbohydrate, Mediterranean, or low-fat diet. N Engl J Med. 2008;359:229–241.
    1. Fraser DA, Thoen J, Bondhus S, Haugen M, Reseland JE, Djøseland O, Førre O, Kjeldsen-Kragh J. Reduction in serum leptin and IGF-1 but preserved T-lymphocyte numbers and activation after a ketogenic diet in rheumatoid arthritis patients. Clin Exp Rheumatol. 2000;18:209–214.
    1. Accurso A, Bernstein RK, Dahlqvist A, Draznin B, Feinman RD, Fine EJ, Gleed A, Jacobs DB, Larson G, Lustig RH, Manninen AH, McFarlane SI, Morrison K, Nielsen JV, Ravnskov U, Roth KS, Silvestre R, Sowers JR, Sundberg R, Volek JS, Westman EC, Wood RJ, Wortman J, Vernon MC. Dietary carbohydrate restriction in type 2 diabetes mellitus and metabolic syndrome: time for a critical appraisal. Nutr Metab (Lond) 2008;5:9.
    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. Schmidt M, Pfetzer N, Schwab M, Strauss I, Kämmerer U. Effects of a ketogenic diet on the quality of life in 16 patients with advanced cancer: A pilot trial. Nutr Metab (Lond) 2011;8:54.
    1. Fine EJ, Segal-Isaacson CJ, Feinman RD, Herszkopf S, Romano MC, Tomuta N, Bontempo AF, Negassa A, Sparano JA. Targeting insulin inhibition as a metabolic therapy in advanced cancer: a pilot safety and feasibility dietary trial in 10 patients. Nutrition. 2012;28:1028–1035.
    1. Macdonald DR, Cascino TL, Schold SCJ, Cairncross JG. Response criteria for phase II studies of supratentorial malignant glioma. J Clin Oncol. 1990;8:1277–1280.
    1. Mueller-Klieser W, Walenta S. Geographical mapping of metabolites in biological tissue with quantitative bioluminescence and single photon imaging. Histochem J. 1993;25:407–420.
    1. Maurer GD, Brucker DP, Bähr O, Harter PN, Hattingen E, Walenta S, Mueller-Klieser W, Steinbach JP, Rieger J. 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.
    1. Chang HT, Olson LK, Schwartz KA. Ketolytic and glycolytic enzymatic expression profiles in malignant gliomas: implication for ketogenic diet therapy. Nutr Metab (Lond) 2013;10:47.
    1. Steinbach JP, Wolburg H, Klumpp A, Probst H, Weller M. Hypoxia-induced cell death in human malignant glioma cells: energy deprivation promotes decoupling of mitochondrial cytochrome c release from caspase processing and necrotic cell death. Cell Death Differ. 2003;10:823–832.
    1. Rieger J, Bähr O, Müller K, Franz K, Steinbach J, Hattingen E. Bevacizumab-induced diffusion-restricted lesions in malignant glioma patients. J Neurooncol. 2010;99:49–56.
    1. de Groot JF, Fuller G, Kumar AJ, Piao Y, Eterovic K, Ji Y, Conrad CA. Tumor invasion after treatment of glioblastoma with bevacizumab: radiographic and pathologic correlation in humans and mice. Neuro Oncol. 2010;12:233–242.
    1. DeLay M, Jahangiri A, Carbonell WS, Hu Y, Tsao S, Tom MW, Paquette J, Tokuyasu TA, Aghi MK. Microarray analysis verifies two distinct phenotypes of glioblastomas resistant to antiangiogenic therapy. Clin Cancer Res. 2012;18:2930–2942.
    1. Hattingen E, Jurcoane A, Bähr O, Rieger J, Magerkurth J, Anti S, Steinbach JP, Pilatus U. Bevacizumab impairs oxidative energy metabolism and shows antitumoral effects in recurrent glioblastomas: a 31P/1H MRSI and quantitative magnetic resonance imaging study. Neuro Oncol. 2011;13:1349–1363.
    1. Tamura H, Hatazawa J, Toyoshima H, Shimosegawa E, Okudera T. Detection of deoxygenation-related signal change in acute ischemic stroke patients by T2*-weighted magnetic resonance imaging. Stroke. 2002;33:967–971.
    1. Seiler A, Jurcoane A, Magerkurth J, Wagner M, Hattingen E, Deichmann R, Neumann-Haefelin T, Singer OC. T2’ imaging within perfusion-restricted tissue in high-grade occlusive carotid disease. Stroke. 2012;43:1831–1836.
    1. Otto C, Kaemmerer U, Illert B, Muehling B, Pfetzer N, Wittig R, Voelker HU, Thiede A, Coy JF. Growth of human gastric cancer cells in nude mice is delayed by a ketogenic diet supplemented with omega-3 fatty acids and medium-chain triglycerides. BMC Cancer. 2008;8:122.
    1. Liu AG, Most MM, Brashear MM, Johnson WD, Cefalu WT, Greenway FL. Reducing the glycemic index or carbohydrate content of mixed meals reduces postprandial glycemia and insulinemia over the entire day but does not affect satiety. Diabetes Care. 2012;35:1633–1637.
    1. Mukherjee P, Mulrooney TJ, Marsh J, Blair D, Chiles TC, Seyfried TN. Differential effects of energy stress on AMPK phosphorylation and apoptosis in experimental brain tumor and normal brain. Mol Cancer. 2008;7:37.
    1. Sarkar NH, Fernandes G, Telang NT, Kourides IA, Good RA. Low-calorie diet prevents the development of mammary tumors in C3H mice and reduces circulating prolactin level, murine mammary tumor virus expression, and proliferation of mammary alveolar cells. Proc Natl Acad Sci USA. 1982;79:7758–7762.
    1. Giovanella BC, Shepard RC, Stehlin JS, Venditti JM, Abbott BJ. Calorie restriction: effect on growth of human tumors heterotransplanted in nude mice. J Natl Cancer Inst. 1982;68:249–257.
    1. Kalaany NY, Sabatini DM. Tumours with PI3K activation are resistant to dietary restriction. Nature. 2009;458:725–731.
    1. Lee C, Raffaghello L, Brandhorst S, Safdie FM, Bianchi G, Martin-Montalvo A, Pistoia V, Wei M, Hwang S, Merlino A, Emionite L, de Cabo R, Longo VD. Fasting cycles retard growth of tumors and sensitize a range of cancer cell types to chemotherapy. Sci Transl Med. 2012;4:124ra27.
    1. Abdelwahab MG, Fenton KE, Preul MC, Rho JM, Lynch A, Stafford P, Scheck AC. The ketogenic diet is an effective adjuvant to radiation therapy for the treatment of malignant glioma. PLoS ONE. 2012;7:e36197.
    1. Zuccoli G, Marcello N, Pisanello A, Servadei F, Vaccaro S, Mukherjee P, Seyfried TN. Metabolic management of glioblastoma multiforme using standard therapy together with a restricted ketogenic diet: case report. Nutr Metab (Lond) 2010;7:33.

Source: PubMed

3
Abonneren