+ All Categories
Home > Documents > A Supplemented High-Fat Low-Carbohydrate Diet for the ... · Purpose:Dysregulated energetics...

A Supplemented High-Fat Low-Carbohydrate Diet for the ... · Purpose:Dysregulated energetics...

Date post: 22-Jul-2020
Category:
Upload: others
View: 0 times
Download: 0 times
Share this document with a friend
15
Cancer Therapy: Preclinical A Supplemented High-Fat Low-Carbohydrate Diet for the Treatment of Glioblastoma Regina T. Martuscello 1,2 , Vinata Vedam-Mai 1,3 , David J. McCarthy 1 , Michael E. Schmoll 1 , Musa A. Jundi 1 , Christopher D. Louviere 1 , Benjamin G. Grifth 1 , Colby L. Skinner 1 , Oleg Suslov 1 , Loic P. Deleyrolle 1 , and Brent A. Reynolds 1,2 Abstract Purpose: Dysregulated energetics coupled with uncontrolled proliferation has become a hallmark of cancer, leading to increased interest in metabolic therapies. Glioblastoma (GB) is highly malignant, very metabolically active, and typically resistant to current therapies. Dietary treatment options based on glucose deprivation have been explored using a restrictive ketogenic diet (KD), with positive anticancer reports. However, negative side effects and a lack of palatability make the KD difcult to implement in an adult population. Hence, we developed a less stringent, supplemented high-fat low-carbo- hydrate (sHFLC) diet that mimics the metabolic and antitumor effects of the KD, maintains a stable nutritional prole, and presents an alternative clinical option for diverse patient populations. Experimental Design: The dietary paradigm was tested in vitro and in vivo, utilizing multiple patient-derived gliomasphere lines. Cellular proliferation, clonogenic frequency, and tumor stem cell population effects were determined in vitro using the neurosphere assay (NSA). Antitumor efcacy was tested in vivo in preclinical xenograft models and mechanistic regulation via the mTOR pathway was explored. Results: Reducing glucose in vitro to physiologic levels, coupled with ketone supplementation, inhibits proliferation of GB cells and reduces tumor stem cell expansion. In vivo, while maintaining animal health, the sHFLC diet signicantly reduces the growth of tumor cells in a subcutaneous model of tumor progression and increases survival in an orthotopic xenograft model. Dietary- mediated anticancer effects correlate with the reduction of mTOR effector expression. Conclusions: We demonstrate that the sHFLC diet is a viable treatment alternative to the KD, and should be considered for clinical testing. Clin Cancer Res; 22(10); 248295. Ó2015 AACR. Introduction Glioblastoma (GB) is the most common high-grade glioma in adults, with extremely poor prognosis. Tentacle-like projections and pseudopalisading necrosis integrate into normal brain tissue making complete surgical resection difcult. Extreme cellular heterogeneity, extensive genetic aberrations, and inadequate early detection make effective long-term control of GB challenging. Current standard of care (SOC) is limited to surgery, radiation, and chemotherapy (temozolomide/TMZ), with median survival of 9 to 12 months and 5-year survival less than 5% (1). Glucose metabolism and the Warburg Effect have gained traction as a potential tumor weakness and exploitable treatment area. Where normal cells utilize glucose for high-yield energy production in the mitochondria (1:36ATP), tumor cells demand higher levels of glucose for diminished energy production, via lactate in the cytosol (1:4ATP) and nucleotide synthesis in the pentose phosphate pathway. This metabolic characteristic, termed the Warburg Effect, is an essential byproduct of rapid cellular proliferation and promoted during tumorigenesis by oncogenic metabolic reprogramming. Hence tumor cells acquire the ability to sustain proliferative signaling mechanisms, which subsequently promotes malignant glycolysis (2). The PI3K/Akt/ mTOR pathway plays a central role in human cancers by way of constitutive activity, increased growth factor expression and gly- colytic activation, resulting in feed-forward loops of glucose consumption, proliferation and survival (3). PTEN, a key regu- lator of the PI3K/Akt/mTOR pathway, is frequently inactivated in GB. Research into dysregulated cellular metabolism has given rise to the notion that dietary therapies for cancer patients may have signicant clinical utility. GB has been proposed to be a promising candidate for dietary intervention due to its substantial reliance and utilization of glucose (4, 5). At the forefront of dietary anticancer therapy is the ketogenic diet (KD), which is a high- fat, low-carbohydrate, low-protein diet, used for decades to treat refractory epileptic seizures. Extreme carbohydrate restriction mimics a fasting state, resulting in reduction of blood glucose and induction of ketone bodies (e.g., b-hydroxybutyrate/BHB; 1 Department of Neurosurgery, College of Medicine, University of Flor- ida, Gainesville, Florida. 2 Interdisciplinary Program in Biomedical Sciences, Neuroscience, College of Medicine, University of Florida, Gainesville, Florida. 3 Center for Movement Disorders and Neuro-res- toration, University of Florida, Gainesville, Florida. Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). Corresponding Authors: Brent A. Reynolds, Department of Neurosurgery, McKnight Brain Institute, 1149 S. Newell Drive, L2-100, Gainesville, FL 32611. Phone: 352-273-8476; Fax: 352-392-8413; E-mail: [email protected]; Loic P. Deleyrolle, Department of Neurosurgery, McKnight Brain Institute, 1149 S. Newell Drive, L2-100, Gainesville, FL 32611. Phone: 352-273-8583; Fax: 352-392- 8413; E-mail: [email protected].edu doi: 10.1158/1078-0432.CCR-15-0916 Ó2015 American Association for Cancer Research. Clinical Cancer Research Clin Cancer Res; 22(10) May 15, 2016 2482 Research. on September 18, 2020. © 2016 American Association for Cancer clincancerres.aacrjournals.org Downloaded from Published OnlineFirst December 2, 2015; DOI: 10.1158/1078-0432.CCR-15-0916
Transcript
Page 1: A Supplemented High-Fat Low-Carbohydrate Diet for the ... · Purpose:Dysregulated energetics coupled with uncontrolled proliferation has become a hallmark of cancer, leading to increased

Cancer Therapy: Preclinical

A Supplemented High-Fat Low-CarbohydrateDiet for the Treatment of GlioblastomaRegina T. Martuscello1,2, Vinata Vedam-Mai1,3, David J. McCarthy1, Michael E. Schmoll1,Musa A. Jundi1, Christopher D. Louviere1, Benjamin G. Griffith1, Colby L. Skinner1,Oleg Suslov1, Loic P. Deleyrolle1, and Brent A. Reynolds1,2

Abstract

Purpose:Dysregulated energetics coupled with uncontrolledproliferation has become a hallmark of cancer, leading toincreased interest in metabolic therapies. Glioblastoma (GB)is highly malignant, very metabolically active, and typicallyresistant to current therapies. Dietary treatment options basedon glucose deprivation have been explored using a restrictiveketogenic diet (KD), with positive anticancer reports. However,negative side effects and a lack of palatability make the KDdifficult to implement in an adult population. Hence, wedeveloped a less stringent, supplemented high-fat low-carbo-hydrate (sHFLC) diet that mimics the metabolic and antitumoreffects of the KD, maintains a stable nutritional profile, andpresents an alternative clinical option for diverse patientpopulations.

Experimental Design: The dietary paradigm was tested in vitroand in vivo, utilizing multiple patient-derived gliomasphere lines.

Cellular proliferation, clonogenic frequency, and tumor stem cellpopulation effects were determined in vitro using the neurosphereassay (NSA). Antitumor efficacy was tested in vivo in preclinicalxenograft models and mechanistic regulation via the mTORpathway was explored.

Results:Reducing glucose in vitro to physiologic levels, coupledwith ketone supplementation, inhibits proliferation of GB cellsand reduces tumor stem cell expansion. In vivo, whilemaintaininganimal health, the sHFLC diet significantly reduces the growth oftumor cells in a subcutaneous model of tumor progression andincreases survival in an orthotopic xenograft model. Dietary-mediated anticancer effects correlate with the reduction of mTOReffector expression.

Conclusions: We demonstrate that the sHFLC diet is a viabletreatment alternative to the KD, and should be considered forclinical testing. Clin Cancer Res; 22(10); 2482–95. �2015 AACR.

IntroductionGlioblastoma (GB) is the most common high-grade glioma in

adults, with extremely poor prognosis. Tentacle-like projectionsand pseudopalisading necrosis integrate into normal brain tissuemaking complete surgical resection difficult. Extreme cellularheterogeneity, extensive genetic aberrations, and inadequate earlydetection make effective long-term control of GB challenging.Current standard of care (SOC) is limited to surgery, radiation,and chemotherapy (temozolomide/TMZ), with median survivalof 9 to 12 months and 5-year survival less than 5% (1).

Glucose metabolism and the Warburg Effect have gainedtraction as a potential tumor weakness and exploitable treatmentarea. Where normal cells utilize glucose for high-yield energyproduction in the mitochondria (1:36ATP), tumor cells demandhigher levels of glucose for diminished energy production, vialactate in the cytosol (1:4ATP) and nucleotide synthesis in thepentose phosphate pathway. This metabolic characteristic,termed the Warburg Effect, is an essential byproduct of rapidcellular proliferation and promoted during tumorigenesis byoncogenic metabolic reprogramming. Hence tumor cells acquirethe ability to sustain proliferative signaling mechanisms, whichsubsequently promotes malignant glycolysis (2). The PI3K/Akt/mTOR pathway plays a central role in human cancers by way ofconstitutive activity, increased growth factor expression and gly-colytic activation, resulting in feed-forward loops of glucoseconsumption, proliferation and survival (3). PTEN, a key regu-lator of the PI3K/Akt/mTOR pathway, is frequently inactivated inGB.

Research into dysregulated cellular metabolism has given riseto the notion that dietary therapies for cancer patients may havesignificant clinical utility. GBhas beenproposed tobe apromisingcandidate for dietary intervention due to its substantial relianceand utilization of glucose (4, 5). At the forefront of dietaryanticancer therapy is the ketogenic diet (KD), which is a high-fat, low-carbohydrate, low-protein diet, used for decades to treatrefractory epileptic seizures. Extreme carbohydrate restrictionmimics a fasting state, resulting in reduction of blood glucoseand induction of ketone bodies (e.g., b-hydroxybutyrate/BHB;

1Department of Neurosurgery, College of Medicine, University of Flor-ida, Gainesville, Florida. 2Interdisciplinary Program in BiomedicalSciences, Neuroscience, College of Medicine, University of Florida,Gainesville, Florida. 3Center for Movement Disorders and Neuro-res-toration, University of Florida, Gainesville, Florida.

Note: Supplementary data for this article are available at Clinical CancerResearch Online (http://clincancerres.aacrjournals.org/).

Corresponding Authors: Brent A. Reynolds, Department of Neurosurgery,McKnight Brain Institute, 1149 S. Newell Drive, L2-100, Gainesville, FL 32611.Phone: 352-273-8476; Fax: 352-392-8413; E-mail: [email protected]; LoicP. Deleyrolle, Department of Neurosurgery, McKnight Brain Institute, 1149 S.Newell Drive, L2-100, Gainesville, FL 32611. Phone: 352-273-8583; Fax: 352-392-8413; E-mail: [email protected]

doi: 10.1158/1078-0432.CCR-15-0916

�2015 American Association for Cancer Research.

ClinicalCancerResearch

Clin Cancer Res; 22(10) May 15, 20162482

Research. on September 18, 2020. © 2016 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 2, 2015; DOI: 10.1158/1078-0432.CCR-15-0916

Page 2: A Supplemented High-Fat Low-Carbohydrate Diet for the ... · Purpose:Dysregulated energetics coupled with uncontrolled proliferation has become a hallmark of cancer, leading to increased

ref. 6). Ketone bodies are suitable energy replacements for normalcellswith functionalmitochondria (7), but have been shown tobeunsuitable for tumor cells (8,9), as tumor cell mitochondrialfunctions are dysregulated (10). Existing preclinical data supportthe KD (11–13) and a calorie-restricted KD (RKD; refs. 14, 15) inthe treatment of brain cancer by diminishing tumor growth andincreasing animal survival. Clinical reports (5), case reports(16,17), and pilot trials (18–20) have demonstrated that the KDis safe, has low toxicity, and is applicable to cancer patients.

These publications note that there are obstacles in its use as acancer treatment due to undesirability for long-term use, withsome reporting mild-to-severe side effects (21). It has beenproposed that more endurable dietary regimens should be devel-oped and tested, as not all patients may tolerate the same dietaryrestrictions (22). Therefore, we sought to develop a less restrictiveKD-like diet that would exhibit the same physiologic phenotypeand antitumor efficacy. By supplementing a high-fat, low-carbo-hydrate (sHFLC), moderate protein diet with specialized medi-um-chain triglycerides [MCT; 60%(30%):30%:10%::Fat(MCT):Protein:Carb], we hypothesize that a more balanced diet can beimplemented, resulting in diminished tumor progression. MCTswere specifically chosen based on carbon chain lengths (C8:C10::97%:3%), which allow them to rapidly diffuse from thegastrointestinal tract into the hepatic portal system and traveldirectly to the liver where they are converted into ketone bodies(6). We believe it is possible to provide a more nutritionallycomplete, flexible, and palatable anticancer diet with the sHFLC,which could target a diverse patient population and increasepatient compliance.

Materials and Methods1

CellsPatient glioblastoma tumors were dissociated and cultured as

previously published (23). All lines tested and authenticatedvia STR-profiling following ICLAC protocols. Culture mediaare serum free, supplemented with EGF at 20 ng/mL. Glucose

restrictedmedia were tested with a glucometer and supplementedwith glucose at plating and every-other-day for the 7-day passagecycle. BHB was administered in themedia at plating day only andmeasured at day 1 andpassaging day 7using theOneTouchmeter.

ImagesAfter sphere formation, cells were fixed with PFA, stained with

DAPI, and imaged using Leica DM 2000 2.5� fluorescent micro-scope. Images were quantified using Macnification.

AnimalsAll experiments were approved by IACUC.NOD/SCID animals

were housed under standard animal husbandry procedures.Transplants: Animals were transplanted per previously publishedprotocols (24). For subcutaneous xenografts, dissociated glioma-spheres were transplanted (1�106). Endpoint criteria of 15�15mm (1766mm3) were used. For intracranial xenografts, animalswere stereotactically injected with dissociated gliomaspheres(2�105) into the striatum (2 ML, and 2.5 DV). Endpoint criteriawere >20% body weight loss, BCS �2, and neurologic deficits.

Diet creation and deliveryThe ketogenic diet was readymade Bioserv #F3666. The sHFLC

and control diets were created using ingredients from Purina TestDiet. The sHFLC diet final caloric% was 10:30:30(30) carbohy-drates:protein:fat(MCTs; 3.2:2.8:1 fats:protein:carb). The sHFLCdiet provided 5.66 kcal/g gross energy where fat, carbohydrates,protein, and fiber comprised 404 g/kg, 125 g/kg, 355 g/kg, 73.12g/kg, respectively. The control diet caloric% was 55:20:25 carbo-hydrates:protein:fat (1:1.8:4.9 fats:protein:carb). The control dietdelivered 4.67 kcal/g gross energy where fat, carbohydrates, pro-tein, and fiber comprised 115 g/kg, 570 g/kg, 208 g/kg, 72 g/kg,respectively. Food (ad libitum)was changed daily. Upon SC tumorpalpation, (1�1mm) animals were randomized and assignedrespective diets. Intracranial animalswere randomized post-trans-plant and assigned respective diets 5 days after final transplant.

Tumor occupancyQuantification of tumor occupancy was determined using

ImageJ. Three images for each brain (4 control, 3 sHFLC, 3 KD)were taken from rostral, medial, and caudal positions of thetumor.

IHCTissue was formaldehyde fixed, cryopreserved, or embedded in

paraffin. Sections were blocked and incubated in primary anti-body overnight at 4�C. Fluorescent secondary antibody wasapplied for 1 hour at RT. Slides were coverslipped-using Vecta-Shield with DAPI. Biotinylated secondary antibody was appliedfor 2 hours at RT followed by 1 hour Vectastain Elite ABC kit andDAB peroxidase kit. Slides were coverslipped with Cytoseal.

Western blot analysisProteins were extracted from subcutaneous tumors and quan-

tified using Qubit. Equal amounts of protein were loaded intoeach well (20–40 mg). Gels were run for 30 minutes at 200 V andthen transferred using a xCell-II blot module (1 hour, 35A, RT).Proteins of interest were detected using chemoluminescense.Membranes were exposed (Kodak), stripped, and probed foractin.1For detailed experimental procedures please see supplemental methods file.

Translational Relevance

The emerging role of metabolism in many types of cancersprovides a novel target for new therapeutics. A possibility isdietary therapies that are designed to take advantage of theunique energetic needs of tumor cells. One such option is theuse of a very high-fat and low-carbohydrate diet (90%:5%,respectively) called theKetogenicDiet (KD). This diet has beenused to treat epilepsy for nearly 90 years, and recently itsapplication as a cancer therapeutic has been explored.Although preclinical studies demonstrate its efficacy and clin-ical case reports support its feasibility, the KD is difficult toimplement due to its stringent nature. Here, we report thedevelopment of an sHFLC, which mimics the main physio-logic effects of the KD, namely reduced glucose and increasedketones. In addition, the sHFLC diet is able to reduce glio-blastoma (GB) tumor cell proliferation and extend lifespan inGB animal models.

Supp. High-fat Low-Carbohydrate Diet for Glioblastoma

www.aacrjournals.org Clin Cancer Res; 22(10) May 15, 2016 2483

Research. on September 18, 2020. © 2016 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 2, 2015; DOI: 10.1158/1078-0432.CCR-15-0916

Page 3: A Supplemented High-Fat Low-Carbohydrate Diet for the ... · Purpose:Dysregulated energetics coupled with uncontrolled proliferation has become a hallmark of cancer, leading to increased

Flow cytometryCells from culture and ex-vivo cells from SC tumors were run on

a BD Biosciences LSRII flow cytometer. Briefly, 0.5�106 cells wereblocked, permeabilized, and incubated with primary antibodiesovernight at 4�C. Secondary antibody was applied for 1 hour atRT. Flow analysis was performed using FlowJo software.

RNA isolation and RT2 PCR Human mTOR ProfilerTotal RNAwas isolated fromSC tumors usingRNeasy PlusMini

RNA isolation kit. RT2first strands andRT2 SYBRGreenROXqPCRMastermix were utilized. RT-PCR was performed using Qiagen(PAHS-098ZE-4), on ABI 7900HT. Housekeeping genes weredetermined using NormFinder.

ResultsRestricted glucose slows proliferation of patient-derivedgliomaspheres

Using four different previously published GB patient-derivedcell lines (L0, L1, L2, and S3; refs. 24–26), we examined the effectsof glucose restriction in-vitro. For this publication, L0will be usedas the primary cell line due to its robust proliferative activity, andL2 will be used as the secondary line since it closely resembles theproliferative and metabolic activity of L0. Cells were grownunder three glucose conditions: normal (glucose) media (NG:500þmg/dL), physiologic (glucose) media (PG: 90–110mg/dL),and physiologic-low (glucose) media (LG: 65–80 mg/dL). Figure1A shows altering glucose concentrations in the media signifi-cantly reduced L0 cell proliferation over time. The cell linesrevealed a significant reduction in mean expansion under PGand LGwhen compared withNG (Supplementary Fig. S1A–S1C),as well as a significant difference between PG and LG media (Fig.1B), as confirmed by a MTT Assay (Fig. 1C). To assess thereduction in fold expansion, L0 was analyzed for proliferation(Ki-67-Fig. 1D; MCM2-Fig. 1E) and apoptosis markers (active-caspase3-Fig. 1F). Lowering glucose concentrations resulted in asignificant reduction in Ki-67 and MCM2 expression, in both PGandLG, aswell as a significant increase in active caspase-3betweenNG and LG. Therefore, alterations in glucose availability, to levelsequivalent to a low glucose state, are sufficient to slow theproliferation of gliomaspheres while concomitantly increasingapoptosis.

The clonogenic frequency and proliferative rate of individualclones (24) were assessed in 96-well culture plates under allconditions (Fig. 1G). Therewas a statistically significant reductionin both the clonogenic frequency (Fig. 1H) and the mean prolif-erative rate of each clone (Fig. 1I), in all lines (Supplementary Fig.S1D—S1I). Quantification of the self-renewing stem cell sym-metrical division rate (KLL; ref. 24) demonstrated a significantdecrease in cancer stem cell expansion under reduced glucoseconditions (Fig. 1J). The combination of diminished stem celldivision rates, cellular fold expansion, and proliferation markersindicates that lowering glucose affects not only the putative stemcell population, but also the non-stem cell population.

Ketone treatment in vitro further suppresses proliferation ofpatient-derived GB cells

To determine ketone effects in vitro and model the sHFLCdietary phenotype (low glucose and high ketones), a doseresponse experiment was performed. BHB treatment in allglucose conditions resulted in a dose-dependent reduction in

fold expansion beginning at 4 mmol/L (Fig. 2A). BHB EC50s werecalculated for each glucose condition (Fig. 2A).On the basis of theBHB EC50 of L0 from PG, as well as the knowledge that this is anachievable, efficacious ketosis plasma concentration for patientson a high-fat diet (7), all in vitro experiments were performedwith4 mmol/L BHB (KTX). The MTT assay also showed reduction inenzymatic activity post BHB treatment beginning at 1mmol/L, butwithout dose dependence (Supplementary Fig. S2A—S2C). Toconfirm the reduction in fold expansion, we performed 50 con-secutive single passages with 3-glucose concentrations with KTX,and we still observed a significant reduction in fold expansion(Fig. 2B) and MTT enzyme activity (Fig. 2C) in all lines (Supple-mentary Fig. S2D–S2E). We assessed proliferation and apoptosisin L0 KTX and found an enhanced reduction in Ki-67 (Fig. 2D)andMCM2 (Fig. 2E) expression, but no increase in active caspase-3 activity (Fig. 2F) post KTX. Clonogenic frequency, proliferationrate of individual clones, and tumor stem cell division rate wereeach determined under restricted glucose, and in conjunctionwith KTX (Fig. 2G). A significant reduction was observed inclonogenic frequency (Fig. 2H) and mean proliferation rate ofeach clone (Fig. 2I) in each glucose media when exposed to KTX,in all lines (Supplementary Fig. S2G–S2L). KLL quantificationdemonstrated a significant reduction in stem cell division rate inall KTX groups compared with glucose controls, with a significantreduction in the low glucose and KTX combination (Fig. 2J) in alllines (Supplementary Fig. S2H, S2J, and S2L]. Together, thisevidence supports the potential anti-tumorigenic effect of thesHFLC dietary phenotype by diminishing the proliferative poten-tial of the putative stem cell and non-stem cell populations.

The sHFLC diet is safe and nutritionally completeDietary breakdown for all xenograft experiments is shown in

Supplementary Fig. Table 1. Detailed descriptions of diet creationand dietary timelines are provided in the Supplemental Methods.Animals placed on the sHFLC and KD had a significant reductionin blood glucose, above hypoglycemic levels, compared to con-trols, with no difference between the sHFLC and KD (Fig. 3A andSupplementary Fig. S3A). Blood ketones were significantlyincreased in mice maintained on the sHFLC and KD to a safelevel, with a statistical difference between the groups (Fig. 3B andSupplementary Fig. S3B). To compare blood glucose and ketonelevels among different anticancer dietary therapies, a simpleglucose ketone index (GKI) is used. The GKI is a single number,and can be used both clinically and preclinically, to identify atherapeutic zone (27). On the basis of this formulation, ouraverage calculated GKIs are 24.4 � 7.14, 3.1 � 1.07, and 1.94� 0.67 for the control, sHFLC and KD, respectively. These valuesare comparable with other published preclinical studies reportinga therapeutic effect using a RKD (range of 1.8–5.7; ref. 27). Tocompare nutritional differences and health status of mice onsHFLC and KD, body weight and blood analyses were performed.Patients on RKD report losing approximately 20% of their bodyweight (28), whereas patients on KD report initial body weightfat-loss followed by stabilization (29). Surprisingly, subcutane-ous-L0 mice fed the sHFLC diet had a significant increase in bodyweight compared with the KD and control (Fig. 3C). In otherxenograft experiments, sHFLC-fed mice had a significant increasein bodyweight comparedwith KD-fedmice only (SupplementaryFig. S3C—S3E]. This may be due to increased caloric intake of anutritionally complete, palatable diet, as animals are fed adlibitum. Nutritional insufficiencies from a high-fat diet can place

Martuscello et al.

Clin Cancer Res; 22(10) May 15, 2016 Clinical Cancer Research2484

Research. on September 18, 2020. © 2016 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 2, 2015; DOI: 10.1158/1078-0432.CCR-15-0916

Page 4: A Supplemented High-Fat Low-Carbohydrate Diet for the ... · Purpose:Dysregulated energetics coupled with uncontrolled proliferation has become a hallmark of cancer, leading to increased

strain on the body's vital organs such as the liver, kidneys, or heart(28). Analysis of serum albumin, alkaline phosphatase (ALP),aspartate aminotransferase (AST), alanine transaminase (ALT),and iron levels in tumor-bearing mice fed the sHFLC diet for over

1month (Fig. 3D–H) showed normal levels in all enzymes tested.KD-fed animals had reduced serum albumin levels (Fig. 3D) andincreased serum ALP levels (Fig. 3E), which has been demon-strated in long-term KD-fed mice (30). Analysis of cholesterol,

Figure 1.Glucose restriction slows proliferation of patient-derived gliomaspheres. All gliomaspheres were cultured on a 7-day passage cycle. A, growth curve ofL0 patient-derived gliomaspheres cultured long-term [over a period of 6 passages (6 weeks)] in altered and normal glucose containing medias. Long-term growthcurve expressed in log-based on calculated fold expansion values for each serial passage. Fold expansion (F) is determined by dividing final density by platingdensity. B, average fold expansions of four patient-derived glioma spheres cultured in normal glucose media (NG: 500þ mg/dL), physiologic glucose media(PG: 90–110 mg/dL) and physiologic-low glucose media (LG: 60–85mg/dL) over a single 7-day passage cycle (n¼ 50 single passages). C, MTT enzymatic assay offour patient-derived gliomasphere cultures, expressed as a percent of control (NG - normal glucose media; n ¼ 3 assays, 6 wells per assay). Each line isstatistically comparedwith its control; no comparison between cell lines ismade. D, quantification of percent positive Ki-67 L0 cells grownover a 7-daypassage cyclemeasured by flow cytometry (n ¼ 10). E, quantification of percent positive MCM2 L0 cells grown over a 7-day passage cycle measured by flow cytometry(n¼ 10). F, quantification of percent positive caspase-3 L0 cells over a 7-day passage cyclemeasured by flow cytometry (n¼ 7). Overall one-wayANOVA significant(P ¼ 0.0483), Bonferroni post hoc comparison insignificant. G, single well image from a 96-well plate of L0 gliomaspheres cultured in altered and normal glucosecontaining medias. Gliomaspheres plated 250 cells per well and grown for 7 days, fixed and stained with 4% PFA/.01% DAPI and imaged using a Leica DMI 4000 Bmicroscope. White box in bottom right corner contains an enlarged gliomasphere. H, quantification of 96-well plates using Macnification to determine sphere-forming frequency expressed as a percentage of control [normal glucose containing media (NG)]; (n ¼ 20 wells). I, quantification of 96-well plates usingMacnification for average sphere diameter in um (n>500 spheres, 20wells). J, calculated KLL values. KLL¼ LN(F)/t, whereby F¼ fold expansion, division of cell countat end of passage/cell count at start of passage, and t ¼ time (days in culture). Statistics performed using a one-way ANOVA with Bonferroni post hoc test.� , P < 0.01; �� , P < 0.001; ��� , P < 0.0001 compared with control (NG). #, P < 0.01; ##, P < 0.001; ###, P < 0.001 compared with PG.

Supp. High-fat Low-Carbohydrate Diet for Glioblastoma

www.aacrjournals.org Clin Cancer Res; 22(10) May 15, 2016 2485

Research. on September 18, 2020. © 2016 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 2, 2015; DOI: 10.1158/1078-0432.CCR-15-0916

Page 5: A Supplemented High-Fat Low-Carbohydrate Diet for the ... · Purpose:Dysregulated energetics coupled with uncontrolled proliferation has become a hallmark of cancer, leading to increased

NG15

15L0

L0

L2

L2100806040205.0

2.5

15

Per

cent

pos

itive

sta

inin

g

10

5

0

0.0

100

80

60

40

54321

******

######

^^

***

***

### ###

###

******

####

**

***

#

^#

^^

*

***##

## ##

*** ***

** *

#####

^

***** #

##

^

***

*** ***

***## ##

###

#

^^ ^^^

10

5

0

NG

NGKTX PG

PGTX

LGTXLG

NGNGKTX PGPGKTX

LGKTXLG

NGNGKTX PGPGKTX

LGKTXLGNG

NGKTX PG

PGKTX

LGKTXLG

NG

NGKTX PG

PGKTX

LGKTXLG

NGNGKTX PGPGKTX

LGKTXLG NG

NGKTX PGPGKTX

LGKTXLG

NGNGKTX PGPGKTX

LGKTXLG

10

5

0

100

80

60

40

4 mmol/LKTX L0

100 200

2

1

0

−1

−2

150

100

50

0

50

0

20

0

0 20 40 60

Ketone treatment (mmol)

Ki67

Normal glucose Physiologic glucose Low glucose

MCM2 Caspase 3+ L0FE

G

H I

J

B

C

A

D

Fold

exp

ansi

onP

erce

nt p

ositi

ve s

tain

ing

Fold

exp

ansi

on

MTT

Enz

ymat

ic a

ctiv

ity(p

erce

nt o

f con

trol)

MTT

Enz

ymat

ic a

ctiv

ity(p

erce

nt o

f con

trol)

Sph

ere

form

ing

frequ

ency

(per

cent

of c

ontro

l)

Aver

age

sphe

re d

iam

eter

(µm

)

KII

PG LG

30

20

10

0

Fold

exp

ansi

on

NGNGKTX

PGPGKTX

LGKTX

LG

80

60

40

20

0

Per

cent

pos

itive

sta

inin

gNG

NGKTX PGPGKTX

LGKTXLG

Martuscello et al.

Clin Cancer Res; 22(10) May 15, 2016 Clinical Cancer Research2486

Research. on September 18, 2020. © 2016 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 2, 2015; DOI: 10.1158/1078-0432.CCR-15-0916

Page 6: A Supplemented High-Fat Low-Carbohydrate Diet for the ... · Purpose:Dysregulated energetics coupled with uncontrolled proliferation has become a hallmark of cancer, leading to increased

triglycerides, HDL, LDL, and non-esterified fatty acids (NEFA)were also performed (Fig. 3I-M), as high cholesterol levels are aconcern with a high-fat diet (31). Reports of mice fed the Bioserv-KD describe increases in hepatic triglycerides and fat accumula-tion, together with a reduction in body weight, due to cholinedeficiency combined with low-protein and high-fat content (32).In line with these findings, animals fed the Bioserv-KD showed asignificant increase in cholesterol (Fig. 3I), triglycerides (Fig. 3J),HDL (Fig. 3K), and LDL levels (Fig. 3L). sHFLC-fed animalsshowed statistically significant high levels of HDL only(Fig. 3K). Together, these results indicate that the sHFLC diet isnutritionally safe.

The sHFLC diet slows tumor progression, increases survival,and reduces tumor burden in subcutaneous and orthotopicxenograft models

In our preclinical subcutaneous xenograft model, L0 GB cellswere grown until palpation (1�1 mm), at which time dietarytreatments were introduced. Overall tumor volume for the sHFLCand KD-fed mice were similar, showing a significant reduction intumor progression compared with controls (Fig. 4A and Supple-mentary Fig. S4A]. In addition, compared with control, bothtreatment groups demonstrated a significant delay in time toreach a tumor volumeof 50mm3; underwhich tumor progressionis not considered to have occurred (Fig. 4B). Both treatmentgroups also demonstrated a significant delay in time to reach1,766mm3 (15�15mm:endpoint; Fig. 4C). These results indicateincreased progression-free survival and time to reach endpoint inboth experimental groups. Similar results were obtained with L2,with respect to body weight, tumor progression, and delayedprogression to endpoint (Supplementary Fig. S3C and S4B andS4C]. Overall, the sHFLC diet demonstrated similar antitumorefficacy as compared with the KD, in a subcutaneous xenograftmodel of tumor progression.

We assessed the efficacy of the sHFLCdiet on tumor burden andhost survival using histologic techniques in our intracranialxenograft model. Brains from tumor-bearing animals werestained with H&E and human-specific Nestin and tumor burdenwas confirmed in all three groups (Fig. 4Dand Supplementary Fig.S4D–S4F]. Animals fed the sHFLC and KD demonstrated a 3-fold

reduction in tumor occupancy compared with controls (Fig. 4E).Brains from mice fed the control and sHFLC diets, when stainedwith active caspase-3 and Ki-67, showed no change in apoptosis,but did show a reduction in proliferation (Supplementary Fig.S4G).

It has been proposed that a potential combinatorial treatmentof metformin with carbohydrate restriction could result inenhanced antitumor efficacy (33). Mice fed the KD and sHFLCalone demonstrated a significant increase in survival comparedwith the control-fed mice (Fig. 4E and Supplementary Fig. S4H),statistically equal to metformin alone. In both xenograft models,metformin alone was able to reduce blood glucose, reduce tumorprogression, and increase survival, yet the combination sHFLCdiet andmetformin showed no additive or synergistic effects (Fig.4E; Supplementary Fig. S4I–S4K). These data show a lack ofcombinatorial efficacy, potentially stemming from an overlap-ping mechanism of action. Metformin is a well-characterizedactivator of AMP-activated protein kinase (AMPK), an enzymeresponsible for cellular energy homeostasis and negative regula-tion of themTORpathway. It is possible that the energy restrictionvia the sHFLC is also activating AMPK and, in turn, inactivatingthe mTOR pathway. These data indicate that the sHFLC diet iscapable of increasing animal survival while minimizing tumorburden, is as effective as metformin, and may mechanisticallyoverlap in AMPK-mediated inactivation of the mTOR pathway.

sHFLCnutritional intervention correlates with downregulationof the mTOR pathway in implanted GB cells

To determine the potential mechanistic effects of the sHFLCdiet, we analyzed 86 key genes in the mTOR pathway, throughrtPCR, using an mTOR Profiler Kit. Four control subcutaneoustumors and four sHFLC-fed subcutaneous tumors were analyzed.Overall, there was a global downregulation of mTORC1/mTORC2 genes in sHFLC-fed tumors compared with control(Fig. 5A), with an inverse relationship observed in negativepathway regulators. All significantly down-or-upregulated genesare shown in Fig. 5B and grouped as functional sets. Comparisonof regulatory genes in functional groups indicates that the mostaffected clusters are the positive mTORC1 regulators and thetranslational effectors downstream of mTORC1/2. Inhibition of

Figure 2.Mimicked sHFLC in vitro (low glucose and ketone treatment) further reduces the proliferation of patient-derived gliomaspheres. A, dose response curveof L0 patient-derived gliomaspheres treated with increasing concentrations of ketones (b-hydroxybutyrate, BHB), expressed via fold expansion. Assay performedin normal glucose-containing media, physiologic media, and low glucose media to determine EC50 value and optimal ketone treatment in vitro. The b-hydroxybutyrate (BHB) EC50 for NG¼ 15.937mmol/L, PG¼ 4.063mmol/L, and LG¼ 2.021 mmol/L. B, fold expansion of consecutive single-passage patient-derivedL0 and L2 cells treated with 4 mmol ketones (KTX) in combination with normal (NG) and altered glucose medias (physiological-PG and low-LG). Foldexpansion calculated by dividing final density by plating density. N ¼ 50þ passages. C, MTT enzymatic assay of patient-derived L0 and L2 treated with 4 mmolketones (KTX) in combination with normal (NG) and altered glucose medias (physiologic-PG and low-LG). Dual wavelength subtraction 750 � 595 OD.Expressed as a percentage of control gliomaspheres grown in normalmedia (NG).N¼ 3 assays/12wells per assay. D, quantification of percent positive Ki-67 L0 cellsgrown over a 7-day passage cycle in normal (NG) and altered glucose medias (physiologic-PG and low-LG), exposed to 4 mmol/L b-hydroxybutyratetreatment (KTX) measured by flow cytometry (n ¼ 10). E, quantification of percent positive MCM2 L0 cells grown over a 7-day passage cycle in normal (NG) andaltered glucose medias (physiologic-PG and low-LG), exposed to 4 mmol/L b-hydroxybutyrate treatment (KTX) measured by flow cytometry (n ¼ 10). F,quantification of percent positive active caspase-3 L0 cells grown over a 7-day passage cycle in normal (NG) and altered glucose medias (physiologic-PG andlow-LG), exposed to 4 mmol/L b-hydroxybutyrate treatment (KTX) measured by flow cytometry (n ¼ 10). G, DAPI-stained 96-well images of patient-derivedGBM cell line 0 grown in normal glucose containing media, physiologic media, and low glucose media with and without 4mmol/L ketone treatment. Gliomaspheresplated 250 cells per well and grown for 7 to 10 days, fixed and stained with 4% PFA/.01% DAPI and imaged using a Leica DMI 4000 B microscope. Whitebox surrounding enlarged gliomasphere in bottom right corner. H, quantification of 96-well DAPI-stained images using Macnification for sphere forming frequency,expressed as a percentage of the control (normal glucose grown cells) n ¼ 20 wells. I, quantification of 96-well DAPI stained images using Macnification foraverage sphere diameter (um). n ¼ 200þ cells. J, KLL Mathematical modeling of stem cell divisions, KLL ¼ LN(F)/t, whereby F ¼ fold expansion, division ofcell count at end of passage/ cell count at start of passage, and t ¼ time (days in culture). Statistics performed using a one-way ANOVA with Bonferronipost hoc comparative test. � , P < 0.01; �� , P < 0.001; ��� , P < 0.0001 when compared with NG control. #, P < 0.01; ##, P < 0.001; ###, P < 0.0001 when comparedwith physiologic control (PG). ^, P < 0.01; ^^, P < 0.001; ^^^, P 0.0001 when compared with low control (LG).

Supp. High-fat Low-Carbohydrate Diet for Glioblastoma

www.aacrjournals.org Clin Cancer Res; 22(10) May 15, 2016 2487

Research. on September 18, 2020. © 2016 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 2, 2015; DOI: 10.1158/1078-0432.CCR-15-0916

Page 7: A Supplemented High-Fat Low-Carbohydrate Diet for the ... · Purpose:Dysregulated energetics coupled with uncontrolled proliferation has become a hallmark of cancer, leading to increased

both mTORC1/mTORC2 has been shown to be a potent anti-cancer therapy (34).

To investigate further the negatively affected mTOR effectorgenes, we assessed ribosomal S6, its activator p70S6K and mTORrelease of 4EBP1. In addition, AMPK activation and phosphory-lation of mTOR were assessed, as metformin primarily targetsAMPK. Flow analysis of multiple ex-vivo single-cell–dissociatedsHFLC-fed tumors confirmed a statistically significant reductionin the phosphorylation of ribosomal S-6 (Fig. 6A), the release ofthe translational repressor 4EBP1 (Fig. 6B) and a significantincrease in AMPK activation (Fig. 6C), when compared withcontrol. The proliferation marker MCM2 was also diminished in

all treatments groups (Fig. 6D). Control-fed tumors served aspositive mTOR controls with constitutive activity levels (PTENnegative; ref. 26), and metformin-fed tumors served as negativemTOR controls (AMPK activator; ref. 35. This was confirmedusing IHC analysis of subcutaneous tumors and xenograft mousebrains (Fig. 6E and F), as well as through Western blot analysis ofsubcutaneous tumors (Fig. 6G; targeting the S6 regulator p70S6K,p-mTOR, AMPK, and multiple epitopes of 4EBP1). The phos-phorylation of 4EBP1, and subsequent release of eIF4E, results ininitiation of translation, and modulation of this release directlyaffects tumorigenicity (36). Therefore, we conclude that thesHFLC diet can potentially diminish cellular proliferation

Figure 3.The sHFLC diet reduces blood glucose, increases blood ketones, maintains body weight, and is nutritionally complete. A, peripheral blood glucose levelsnormalized to bodyweight of animals on their respective diets, expressed as arbitrary units (mg/dL per gramof bodyweight). Blood glucose levels obtained using aPrecision Xtra One Touch glucose meter of animals fed control, sHFLC and ketogenic diets. Of note, 5 mL of blood drawn using tail snip method. Bloodglucose values can be found in Supplementary Fig. S3A. B, peripheral blood ketone body levels normalized to body weight of animals on their respective diets,expressed as arbitrary units (mmol per gram of body weight). Blood ketone body levels obtained using a Precision Xtra One Touch ketone meter of animalsfed control, sHFLC and ketogenic diets. Of note, 5 mL of blood drawn using tail snip method. Blood ketone values can be found in Supplementary Fig. SB. C,averaged animal body weight (g) over time. Weighted three times per week post-tumor palpation. D–M, serum blood analytes tested in tumor-bearing animals fedthe control, sHFLC and ketogenic diets. Blood obtained via retro orbital eye bleeds before sacrifice. Blood samples spun and serum separated. ComparativeClinical Pathology Services, LLC, carried out Blood analyte testing. � , P < 0.01; �� , P < 0.001; ��� , P < 0.0001 when comparing with control fed group. #, P < 0.01;##, P < 0.001; ###, P < 0.0001 when compared with the sHFLC; All statistics performed using a one-way or two-way ANOVA with Bonferroni post hoc test.

Martuscello et al.

Clin Cancer Res; 22(10) May 15, 2016 Clinical Cancer Research2488

Research. on September 18, 2020. © 2016 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 2, 2015; DOI: 10.1158/1078-0432.CCR-15-0916

Page 8: A Supplemented High-Fat Low-Carbohydrate Diet for the ... · Purpose:Dysregulated energetics coupled with uncontrolled proliferation has become a hallmark of cancer, leading to increased

through AMPK-mediated mTOR inactivation of 4EBP1 andthrough protein synthesis via ribosomal S6.

DiscussionWe report a dietary intervention that produces low circulating

glucose while elevating ketones and results in a substantialreduction inGB cellular proliferation.Maintaining in-vitro glucoseat levels representing physiologic (90–110mg/dL)or physiologic-lowbutnot hypoglycemic (65–80mg/dL), reduces the clonogenicfrequency of gliomaspheres, potentially affecting the cancerstem cell population. The addition of BHB also reduces GB

proliferation and when combined with reduced glucose levels,results in further reduction in proliferation. Here, we demonstratethat a high-fat, low-carbohydrate diet supplementedwithMCToil(sHFLC) is able to slow tumor progression and increase survival.In vivo, the sHFLC diet was similar to the ketogenic diet (KD) inantitumor efficacy, but showed nutritional advantages in bodyweight, organ enzyme levels, and lipid profile. Finally, we dem-onstrate that the sHFLC diet affects the mTOR signaling pathwayby reducing expression of upstream regulators and translationaldownstream effectors.

We utilized patient-derived GB stem cell lines that have beendemonstrated tonot only recapitulate the genotype andphenotype

2,000A B C

DE

F

Control (n = 6) 30 60

55

50

45

40

35

50

100

80

60

40

20

40 60

Control (n = 11)Metformin (n = 7)sHFLC (n = 10)sHFLC + Met (n = 8)KD (n = 10)

***

*** ***

***

***

80

Days post transplant

0

40

30

20

10

0

30

Tim

e to

reac

h 17

66 m

m3

(day

s)

Pro

gres

sion

-free

sur

viva

l (da

ys)

Tum

or O

ccup

ancy

(%)

Per

cent

sur

viva

l

20

10

0

Contro

l

sHFLC KD

Contro

l

sHFLC KD

Contro

l

sHFLC KD

sHFLC (n = 7)KD (n = 7)

Tum

or v

olum

e (m

m3 )

1,500

1,000

500

010 20 4030

Days post transplant

H&E

Con

trol B

rain

1sH

FLC

Bra

in 1

KD

Bra

in 1

Nestin

Figure 4.sHFLC diet slows tumor progression, increases progression-free survival, and overall survival while reducing tumor burden in a subcutaneous and orthotopicGB xenograft models. A, averaged overall tumor volume of animals with subcutaneous tumors fed either a control, sHFLC or ketogenic diet. Tumor calculationshown in Supplementary Fig. S4A. Tumors measured three times per week using calipers and calculated overall tumor volume averaged across all mice.B, delayed time progression to reach 50mm3 total tumor volume. C, delayed time progression to reach 1,766mm3 (endpoint) total tumor volume. D, H&E andNestin-stained whole brain OCT sections of mice fed a control, sHFLC diet or ketogenic diet (KD). Two control, two sHFLC-fed, and two KD-fed brains are illustrated in thetable. Human GBM cells were identified using anti-human Nestin antibody and visualized using the ABC-Elite peroxidase method (Vector Laboratories).Counterstaining of the nuclei was performed using hematoxylin. Additional anti-human Nestin fluorescent staining showed accompanying the permanent stainingto illustrate cellular invasion. E, analysis of cross-sectional area shows significantly decreased tumor occupancy for mouse brains fed the sHFLC diet andketogenic diet compared with controls. Graph shows average SE over 5 to 8 sections per group. P <0.0001. F, Kaplan–Meier survival curve ofmice transplantedwithL0 patient-derived GB treatedwith a control, sHFLC,metformin, sHFLCþmetformin, or ketogenic diet. � , P <0.01; �� , P <0.001; ��� , P <0.0001when comparingwithcontrol-fed group. Statistics performed for Kaplan–Meier using a Mandel-Cox, Nestin quantification using a Student t test and one-way ANOVA for tumorprogression.

Supp. High-fat Low-Carbohydrate Diet for Glioblastoma

www.aacrjournals.org Clin Cancer Res; 22(10) May 15, 2016 2489

Research. on September 18, 2020. © 2016 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 2, 2015; DOI: 10.1158/1078-0432.CCR-15-0916

Page 9: A Supplemented High-Fat Low-Carbohydrate Diet for the ... · Purpose:Dysregulated energetics coupled with uncontrolled proliferation has become a hallmark of cancer, leading to increased

of the primary tumor (37), but also to be driven by a cancer stemcell population (38). They are grown in serum-free conditions,which serves as a clinically relevant model for studying GB in vitro

(39). Standard tissue culturemedia are supersaturatedwith glucoseto provide an excess of nutrients for continuous growth. However,available glucose is variable and highly dependent on nutritional

MY01C

A

B

CT

1C

T2

CT

3C

T4

TX

1T

X2

TX

3T

X4

PRKCGRPS6KA1

GSK3BPPP2R4

AKT2RPS6KA2

CAB39CAB39L

IRS1TSC1TSC2

RPS6KA5MAPK3STK11IKBKBPDPK1MAPK1PRKCEPIK3CB

TP53RICTOR

EIF4BRPS6KB1RPS6KB2

RRAGCDEPTORPIK3CD

PLD1PLD2

RPT0RPRKCB

CHUKVEGFBDDIT4L

MT0REIF4EBP1

RPS6HIF1A

MLST8ILK

PPP2CARRAGDP1K3C3CDC42

PRKAG2PPP2R2BEIF4EBP2

VEGFAIGF1

RRAGBPRKAG3

INSRFKBP8

RRAGAAKT1

MAPKAP1AKT3

FKBP1AP1K3CAAKT1S1

EIF4EP1K3CG

HSPA4RHEB

PRKCAHRASPTEN

VEGFCPRKAB1

RH0APRKAA1PRKAA2PRKAB2IGFBP3

DDIT4INS

PRKAG1

Figure 5.The sHFLC diet diminishes mTORC1/2 signaling. A,clustergram of 78 selected mTOR-associated genesfrom Qiagen RT2Profiler with 86 key mTOR genes.Color gradient of clustergram shows consistentdownregulation of the mTOR pathway. Clustergramhas four different animal tumors from the control andsHFLC-fed groups. Tumors analyzed weresubcutaneously transplanted patient-derived line 0gliomaspheres. B, table of significant P values ofselected mTORC1/2 substrate genes grouped infunctional sets. Housekeeping geneswere determinedusing NormFinder and normalized using the Qiagenanalysis software. Statistics performed using aStudent t test.

Martuscello et al.

Clin Cancer Res; 22(10) May 15, 2016 Clinical Cancer Research2490

Research. on September 18, 2020. © 2016 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 2, 2015; DOI: 10.1158/1078-0432.CCR-15-0916

Page 10: A Supplemented High-Fat Low-Carbohydrate Diet for the ... · Purpose:Dysregulated energetics coupled with uncontrolled proliferation has become a hallmark of cancer, leading to increased

Figure 6.sHFLC potential mechanism of action. A–D, assessment of mTOR substrate activation via flow cytometry. Graphically represented as a percent of positivestaining based on control. Mean fluorescent intensity for each treatment group was averaged across multiple animal samples and normalized to be expressedas a percent compared with the normal fed control mice¼ 100%. E, immunofluorescent-stained subcutaneous xenograft tumor sections (5 mm) cryopreserved andembedded in OCT. Green ¼ antibody/Blue ¼ DAPI. All images were taken with a 10� lens on a Leica DM 4000 microscope. F, immunofluorescent-stainedmouse brain sections from intracranial xenograft (10�m) cryopreserved and embedded in OCT. Green¼ antibody/Blue¼ DAPI. All images were taken with a 10�lens on a Leica DM 4000 microscope. G, Western blot analysis for mTOR substrates using Metformin treatment as a positive control as well as mTORsiRNA. Quantification with ImageJ to the right. � , P < 0.01; �� , P < 0.001; ���, P < 0.0001 compared with control; #, P < 0.01; ##, P < 0.001; ###, P < 0.0001 comparedwith sHFLC; ^, P < 0.01; ^^, P < 0.001; ^^^, P < 0.0001 compared with KD. All statistics performed using one-way ANOVA with Bonferroni post hoc test.

www.aacrjournals.org Clin Cancer Res; 22(10) May 15, 2016 2491

Supp. High-fat Low-Carbohydrate Diet for Glioblastoma

Research. on September 18, 2020. © 2016 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 2, 2015; DOI: 10.1158/1078-0432.CCR-15-0916

Page 11: A Supplemented High-Fat Low-Carbohydrate Diet for the ... · Purpose:Dysregulated energetics coupled with uncontrolled proliferation has become a hallmark of cancer, leading to increased

status or disease state in vivo. Excess glucose, as seen in GB patientswithpersistenthyperglycemia, leads topoorpatient survival (40). Ithas also been suggested that diets with a high glycemic index mayincrease the risk of tumorigenesis (41), and low-carbohydrate,high-protein diets that limit circulating glucose can delay cancerdevelopment and progression (42). In vitro, glucose withdrawaland treatment of GB cells with the non-metabolizing glucoseanalog 2-deoxy-glucose (2DG) results in up to 95% growth inhi-bition (43). These studies demonstrate the dependence of cancercells on glucose to promote survival and cellular growth. To assessthe reliance of our patient-derived GB lines on glucose, we reducedglucose concentrations. Using four GB lines, we show a significantdecrease in overall proliferation, an increase in apoptosis, andreductions in both clonogenic frequency and stem cell symmetricdivision rate when GBs are placed under glucose restriction (Fig. 1and Supplementary Fig. S1). These findings, together with otherpublished studies, support the hypotheses that high glucose con-tributes to tumor growth and that reducing glucose levels canattenuate tumor cell proliferation.

During periods of fasting or starvation, ketone bodies areproduced in the liver as compensatory energy sources, whereBHB/acetoacetates are metabolized into acetyl-coA and utilizedwithin the TCA cycle for oxidative ATP production (6). In contrastwith normal cells, GB cells have been shown to be incapable ofutilizing ketones as a metabolic substrate, due to a lack of 3-hydroxybutyrate metabolizing enzymes (44) and low expressionlevels of mitochondrial ketolytic enzymes (OXCT1 and BDH1;ref. 45). Neuroblastoma has also shown decreased expression ofsuccinyl-coenzyme A:3-oxoacid coenzyme A transferase (SCOT),a key enzyme in ketone body metabolism (46). In vitro treatmentof glioma cells with acetoacetate or BHB is able to delay both cellgrowth and metabolic enzyme activity (8), and reduces prolifer-ation and cell viability of VM-M3 cells (9). Other reports indicateBHB acts as an endogenous and specific inhibitor of class I histonedeacetylases (HDAC; ref. 47). HDAC inhibitors are an emergingantiproliferative, proapoptotic, and cell-cycle inhibiting antican-cer therapy (48). BHB treatment of HEK293 cells resulted in adose-dependent increase inhistone acetylation, even at 1-2mmol/L. In vivo, 24-hour–fasted or calorie-restricted animals showedincreased histone acetylation with ketone levels between 1.2 to1.5mmol/L (47). In addition, ketone body administration in vitrohas shown regulatory effects on glucose metabolism (49) andglycolysis (8). Therefore, BHB emerges as a novel environmental(dietary) regulator ofmetabolic health, chromatinmodifications,and epigenetic gene regulation (50). In contrast, Lisanti andcolleagues have reported in abreast cancermodel that high ketonelevels can promote tumor progression in-vivo and increasedmigration in vitro, but not in vivo (51). This study raises thepossibility that certain types of cancers, or biologic systems, maybe differentially affected by elevated ketones. Here, we show thattreatment of four GB stem cell lines, with increasing concentra-tions of BHB, caused a dose-dependent inhibition of prolifera-tion,with supra-physiologic concentrations (>40mmol/L), result-ing in almost no proliferation (Fig. 2A). Conversely, BHB treat-ment in escalating doses, as assessed by a MTT assay, indicated athreshold of anti-enzymatic activity, rather than a true doseresponse [Supplementary Fig. S2A—S2C]. We hypothesize thatthe previously discussed metabolic interaction of BHB may yielddifferential effects onGBproliferation and enzymatic activity. TheMTT assay measures NAD(P)H flux within the cell, which directlyrelates to metabolic activity, while cell numbers calculates fold

expansion. These different measurable outcomes indicate BHBtreatment has varying effects, potentially fromHDACor glycolysisinhibition, in cell populations that also differ in proliferation andmetabolic activity (Fig. 1B and C). Treatment of GB stem cell lineswith a constant physiologic concentration of BHB (4mmol/L), asseen in KDpatients, resulted in reduction of clonogenic frequencyand symmetrical stem cell divisions, suggesting that elevatedketones affect the putative cancer stem cell population (24). Thiseffect was seen in all patient-derived cell lines, which include theclassical (L0, L1, L2) and proneural (S3) subtypes of GB. GBsubtypes represent the variation in mutational status that mayaffect some aspect of metabolism and glycolysis. The proneuralsubtype exhibits isocitate dehydrogenase 1 (IDH1) mutations,which alter the cell's ability to regulate redox,NAD(P)H levels andmetabolize several macromolecules (52), potentially making thissubtype more susceptible to metabolic interventions.

The combination of reduced glucose and increased ketonebodies has shown an enhanced anticancer effect. The KD mimicsthese biologic effects and has been proposed as a treatment for GBand other cancers (5). This was first explored in two pediatricpatients with advanced-stage astrocytomas, following SOC treat-ments, and was able to produce long-term (>5 years) tumormaintenance, employing a KD supplemented with MCT oil(16). Since this landmark report, numerous groups have inves-tigated the antitumor efficacy of a KD and an RKD in several typesof cancer. RKD in experimentalmousemodels of glioma has beenshown to be antitumorigenic, antiangiogenic, and prosurvival(14), while also being anti-invasive, anti-inflammatory, andproapoptotic by targeting signaling pathways related to glucoseand glutamine metabolism (4). In animal models, feeding withtheKD ad libitumhas been reported to increase survival and reducetumor growth (11). Other preclinical animal models such asgastric cancer (53), colon cancer (54), and metastatic cancer(13) have used the KD, reporting similar antitumorigenic effects.Several case reports describe the applicability (20) and combina-torial safety of KD and RKDwith current SOC therapies (17), withsome patients experiencing stable disease on the diet and othersexperiencing unable tomaintain (19). In 2014, the first phase oneclinical trial assessing the safety of the KD on recurrent glioblas-toma determined that the diet is safe and feasible, warrantingother trials to explore the combinatorial potential of the KD withcytotoxic treatments (18). The KD in the context of cancer ispromising, however patient populations are diverse, as are theirdietary needs (21), creating a demand for alternative dietaryinterventions for tumor management (22).

We designed the sHFLC diet for long-term sustainable main-tenance of GB and for increased flexibility and palatability. Weutilized NOD/SCID xenografts in this study for the advantages instudying measureable outcomes of human tumor responses totherapy. It should be noted, however, that NOD/SCID micedisplay the effects of both type I and type II diabetes, are immu-nocompromised and have an altered metabolic state. Our pre-clinical models of tumor progression and survival support thefeasibility of the sHFLC diet, as we can effectively slow theprogression of GB and increase animal survival with diet alone(Fig. 4), using two patient-derived GB lines. Notably, sHFLC-fedanimals, compared with the Bioserv-KD, had superior nutritionalstatus demonstrated by increased body weight and improvedblood analyte and lipid profiles (Fig. 3). The Bioserv-KD weutilized contains large amounts of lard-based fat, and low levelsof protein and choline, which increases hepatic triglycerides and

Martuscello et al.

Clin Cancer Res; 22(10) May 15, 2016 Clinical Cancer Research2492

Research. on September 18, 2020. © 2016 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 2, 2015; DOI: 10.1158/1078-0432.CCR-15-0916

Page 12: A Supplemented High-Fat Low-Carbohydrate Diet for the ... · Purpose:Dysregulated energetics coupled with uncontrolled proliferation has become a hallmark of cancer, leading to increased

cholesterol while simultaneously reducing body weight (32).Furthermore, the 1:6 carbohydrate:fat KD utilized in this studyis higher in fat content than the KD typically administered topatients (1:4). Patients on a 1:4 or 1:3 KD can better manage fatsources to attenuate issues such as weight loss and increasedcholesterol, which have been demonstrated here with animalson a 1:6 KD and attenuated with animals on the sHFLC. Our GKIvalues were comparable to previous publications, but were moreindicative of an RKD than an ad libitum KD. However, whencomparing GKI values to human data, patients receiving a KDsupplemented with MCTs (also fed ad libitum) exhibit valuescloser to the ones we report. We hypothesize that the utilizationof MCTs, which inherently increase ketone body production invivo, may skew the GKI towards a more favorable value. MCTsupplementation has also been shown to inhibit proteolytic andlipolytic factors of cachexia and increase body weight in cachecticcancer patients (55), which is not typically a problem for braintumor patients but may still be useful for patients with othercancer types susceptible to metabolic intervention.

Pharmacologic intervention to temper circulating glucose hasbeen explored,with the assumption that a combinationofKDandmetformin could work together as a comprehensive anticancertherapy (33). We proposed to enhance our dietary paradigmwiththe addition of Metformin to the sHFLC diet (SupplementaryTable S1). Metformin alone significantly reduced tumor volumeand enhanced survival in two preclinical glioma models, indicat-ing metformin as a promising anticancer therapy. When metfor-min was combined with the sHFLC diet, there were no additive orsynergistic effects (Supplementary Fig. S4), indicating possibleoverlapping mechanisms of action. Metformin has multiple pro-posed antitumor mechanisms, but it is well characterized toactivate AMPK, a negative regulator of the mTOR pathway(33). The anticonvulsant mechanism of the KD is still unclear,however, one proposed mechanism is also inhibition of themTOR pathway (56, 57). Taking into account the proposed KDmechanism, the known metformin mechanism and the lack ofcombinatorial antitumor efficacy, we posit that a potential mech-anism of action for the sHFLC diet is through diminished mTORsignaling (Figs. 5 and 6). A large body of literature exists on themTOR pathway, its substrates and how its hypo/hyper-activationresults in various disease states (35). ThemTOR pathway is one ofthe largest and most utilized pathways in cellular signaling, withtwo complexes (mTORC1/mTORC2) that have demonstrated arole in tumorigenesis. Recently, it's been shown that inhibition ofboth mTORC1/mTORC2 signaling results in dramaticallyreduced cell viability in glioma cell lines, as well as inhibitionof tumor growth in vivo (34). In our assessment of the sHFLCdiet'seffects on the mTOR pathway, we found significant reduction inboth mTORC1/mTORC2 signaling (Fig. 5). In our preclinicalmodels, sHFLC-fed tumors showed significant reduction indownstream mTOR effector signaling, whereby reduction inphosphorylation of 4EBP1 and ribosomal S6 inhibits both trans-lation and protein synthesis needed for cellular replication (36)(Fig. 6). Taken together, these findings indicate that inhibition ofthe mTORC1/2 pathway can be achieved through dietary inter-vention, resulting in a potent anti-cancer treatment.

Our work demonstrates that there is a distinct relationshipbetween metabolism and proliferation that can be exploited bychanging the energy sources in the body. Further research intothe biochemical reactions of metabolic intermediates may shedmore light on how ketone bodies are differentially utilized bytumor cells, as the role of mitochondria in tumor propagationand carcinogenesis is multifaceted and incompletely under-stood. Nevertheless, we effectively show that a combinationof low glucose and high ketones results in negative proliferativeeffects on gliomaspheres, which can be translated in vivo withthe sHFLC diet. This diet reduces overall tumor burden andincreases survival, equivalent to a strict 1:6 KD, and has acomplete nutritional profile. Hence we propose that dietarytherapy, such as the sHFLC diet, could be utilized in themanagement of GB.

Disclosure of Potential Conflicts of InterestNo potential conflicts of interest were disclosed.

Authors' ContributionsConception and design: R.T. Martuscello, V. Vedam-Mai, C.L. Skinner,L.P. Deleyrolle, B.A. ReynoldsDevelopment of methodology: R.T. Martuscello, C.L. Skinner, O. Suslov,L.P. Deleyrolle, B.A. ReynoldsAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): R.T. Martuscello, D.J. McCarthy, M.E. Schmoll,C.D. Louviere, B. Griffith, C.L. Skinner, B.A. ReynoldsAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): R.T. Martuscello, V. Vedam-Mai, M.A. Jundi,C.D. Louviere, L.P. Deleyrolle, B.A. ReynoldsWriting, review, and/or revision of the manuscript: R.T. Martuscello,V. Vedam-Mai, L.P. Deleyrolle, B.A. ReynoldsAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): R.T. Martuscello, D.J. McCarthy, C.D. Louviere,O. Suslov, L.P. Deleyrolle, B.A. ReynoldsStudy supervision: V. Vedam-Mai, L.P. DeleyrolleOther (Submitted by Regina Martuscello regarding all images in this man-uscript submission): B.A. Reynolds

AcknowledgmentsThe authors thank all members of the B.A.R lab and undergraduate

volunteers for their assistance, specifically, Andrew Naples, James McGui-ness, Lindsey Chason, Hunter Futch, and Gretter Diaz. The authors alsothank their funding sources and the McKnight Brain Institute core facilities offlow cytometry and the cell and tissue analysis core (CTAC), in addition tothe University of Florida College of Medicine and the InterdisciplinaryProgram in Biomedical Sciences, and Barbara Frentzen and the FCBTR forbrain tumor samples.

Grant SupportThis work was financially supported by McKnight Brain Institute, Depart-

ment of Neurosurgery (to B.A. Reynolds), Florida Center for Brain TumorResearch (to B.A. Reynolds), National Brain Tumor Society (to B.A. Reynolds),NIH – NINDS R24 NS086554-01 (to B.A. Reynolds), NIH/NCI - R21CA141020-01 (to B.A. Reynolds), and American Cancer Society Chris DiMarcoInstitutional Research Grant (to L.P. Deleyrolle).

Received April 15, 2015; revised October 30, 2015; accepted November 12,2015; published OnlineFirst December 2, 2015.

References1. Fuentes-Raspall R, Puig-Vives M, Guerra-Prio S, Perez-Bueno F, Marcos-

Gragera R. Population-based survival analyses of central nervous systemtumors from 1994 to 2008. An up-dated study in the temozolomide-era.Cancer Epidemiol 2014;38:244–7.

www.aacrjournals.org Clin Cancer Res; 22(10) May 15, 2016 2493

Supp. High-fat Low-Carbohydrate Diet for Glioblastoma

Research. on September 18, 2020. © 2016 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 2, 2015; DOI: 10.1158/1078-0432.CCR-15-0916

Page 13: A Supplemented High-Fat Low-Carbohydrate Diet for the ... · Purpose:Dysregulated energetics coupled with uncontrolled proliferation has become a hallmark of cancer, leading to increased

2. Ward PS, Thompson CB. Metabolic reprogramming: a cancer hallmarkeven warburg did not anticipate. Cancer Cell 2012;21:297–308.

3. Laplante M, Sabatini DM. mTOR signaling in growth control and disease.Cell 2012;149:274–93.

4. Seyfried TN, Flores R, Poff AM, D'Agostino DP, Mukherjee P. Metabolictherapy: a new paradigm formanagingmalignant brain cancer. Cancer Lett2014;356:289–300.

5. Woolf EC, Scheck AC. The ketogenic diet for the treatment of malignantglioma. J Lipid Res 2015;56:5–10.

6. VanItallie MDTB, Thomas H, Nufert BA. Ketones: metabolism's uglyduckling. Nutr Rev 2003;61:327–41.

7. Veech R. The therapeutic implications of ketone bodies: the effects ofketone bodies in pathological conditions: ketosis, ketogenic diet, redoxstates, insulin resistance, and mitochondrial metabolism. ProstaglandinsLeukot Essent Fatty Acids 2004;70:309–28.

8. Patel MS, Russell JJ, Gershman H. Ketone-body metabolism in glioma andneuroblastoma cells. Proc Natl Acad Sci USA 1981;78:7214–8.

9. Poff AM, Ari1 CP, Arnold TN, Seyfried D'Agostino DP. Ketone supple-mentation decreases tumor cell viability and prolongs survival ofmicewithmetastatic cancer. Int J Cancer 2014;135:1711–20.

10. Macleod KF. Mitochondrial dysfunction in cancer. Fronteirs Oncol 2013;3:1–28.

11. Stafford P, Abdelwahab MG, Kim DY, Preul MC, Rho JM, Scheck AC. Theketogenic diet reverses gene expression patterns and reduces reactiveoxygen species levels when used as an adjuvant therapy for glioma. NutrMetab 2010;7:1–11.

12. Scheck AC, Abdelwahab MG, Fenton KE, Stafford P. The ketogenic diet forthe treatment of glioma: insights from genetic profiling. Epilepsy Res2011;100:327–37.

13. Poff AM, Ari C, Seyfried TN, D'Agostino DP. The ketogenic diet andhyperbaric oxygen therapy prolong survival in mice with systemic meta-static cancer. Plos ONE 2013;8:e65522.

14. ZhouW,Mukherjee P, KiebishMA,MarkisWT,Mantis JG, Seyfried TN. Thecalorically restricted ketogenic diet, an effective alternative therapy formalignant brain cancer. Nutr Metab 2007;4:1–15.

15. Seyfried BTN, Kiebish M, Marsh J, Mukherjee P. Targeting energy metab-olism in brain cancer through calorie restriction and the ketogenic diet.J Cancer Res Ther 2009;5:S7–15.

16. Nebeling L, Floro Miraldi, Shurin S, Lerner E. Effects of a ketogenic diet ontumor metabolism and nutitional status in pediatric oncology patients:two case reports. J Am College Nutr 1995;14:202–8.

17. Zuccoli G, Marcello N, Pisanello A, Servadei F, Vaccaro S, Mukhergee P,et al. Metabolic management of glioblastoma multiforme using standardtherapy together with a restricted ketogenic diet: Case Report. Nutr Metab2010;7:33–40

18. Rieger J, Bahr O, Maurer GD, Hattingen E, Franz K, Brucker D, et al. ERGO:A pilot study of ketogenic diet in recurrent glioblastoma. Int J Oncol 2014;44:1843–52.

19. Schmidt M, Pfetzer N, Schwab M, Strauss I, K€ammerer U. Effects of aketogenic diet on the quality of life in 16 patients with advanced cancer: Apilot trial. Nutr Metab 2011;8:54–67.

20. Champ CE, Palmer JD, Volek JS, Werner-Wasik M, Andrews DW, EvansJJ, et al. Targeting metabolism with a ketogenic diet during thetreatment of glioblastoma multiforme. J Neurooncol 2014;117:125–31

21. Simone BA, Champ CE, Rosenberg AL, Berger AC, Monti DA, Dicker AP,et al. Selectively starving cancer cells through dietary manipulation: meth-ods and clinical implications. Future Oncol 2013;9:959–76.

22. Lv M, Zhu X, Wang H, Wang F, Guan W. Roles of caloric restriction,ketogenic diet and intermittent fasting during initiation, progression andmetastasis of cancer in animal models: a systematic review and meta-analysis. PloS ONE 2014;9:1–17.

23. Azari H, Millette S, Ansari S, Rahman M, Deleyrolle LP, Reynolds BA.Isolation and expansion of human glioblastoma multiforme tumor cellsusing the neurosphere assay. J Visualized Exp 2011;56:1.

24. Deleyrolle LP, Ericksson G, Morrison BJ, Lopez JA, Burrage K, Burrage P,et al. Determination of somatic and cancer stem cell self-renewing sym-metric division rate using sphere assays. PLoS ONE 2011;6:e15844.

25. Deleyrolle LP, Harding A, Cato K, Siebzehnrubl FA, Rahman M, Azari H,et al. Evidence for label-retaining tumour-initiating cells in human glio-blastoma. Brain 2011;134:1331–43.

26. Siebzehnrubl FA, Silver DJ, Tugertimur B, Deleyrolle LP, Siebzehnrubl D,Sarkisian MR, et al. The ZEB1 pathway links glioblastoma initiation,invasion and chemoresistance. EMBO Mol Med 2013;5:1196–212.

27. Meidenbauer JJ, Purna Mukherjee, Seyfried TN. The glucose ketone indexcalculator: a simple tool to monitor therapeutic efficacy for metabolicmanagement of brain cancer. Nutr Metab 2015;12:1–7.

28. Dirks AJ, Leeuwenburgh C. Caloric restriction in humans: Potential pitfallsand health concerns. Mech Ageing Dev 2005;127:1–7.

29. Wibisono C, Rowe N, Beavis E, Kepreotes H, Mackie FE, Lawson JA, et al.Ten-year single-center experience of the ketogenic diet: factors influencingefficacy, tolerability, and compliance. J Pediatr 2015;166:1030–6

30. Garbow JR, Doherty JM, Schugar RC, Travers S, Weber ML, Wentz AE, et al.Hepatic steatosis, inflammation, and ER stress in mice maintained longterm on a very low-carbohydrate ketogenic diet. Am J Physiol GastrointestLiver Physiol 2011;300:G956–67.

31. Kwiterovich PJr, Vining E, Pysik P, Skolasky RJr, Freeman J. Effects of a high-fat ketogenic diet on plasma levels of lipids, lipoproteins, and apolipo-proteins in children. JAMA 2003;290:912–20.

32. Schugar RC, Xiaojing Huang, Moll Ashley R, Brunt Elizabeth M, CrawfordPA. Role of choline deficiency in the Fatty liver phenotype ofmice fed a lowprotein, very low carbohydrate ketogenic diet. PLoS ONE 2013;8:e74806–e74806.

33. Oleksyszyn J. The complete control of glucose level utilizing the compo-sition of ketogenic diet with the gluconeogenesis inhibitor, the anti-diabetic drug metformin, as a potential anti-cancer therapy. Med Hypoth-eses 2011;77:171–3.

34. Luchman HA, Stechishin ODM, Nguyen SA, Lina XQ, Cairncross G, WeissS. Dual mTORC1/2 Blockade inhibits glioblastoma brain tumor initiatingcells in vitro and in vivo and synergizes with temozolomide to increaseorthotopic xenograft survival. Clin Cancer Res 2014;20:5756–67.

35. Alayev A,HolzMK.mTORsignaling for biological control and cancer. J CellPhysiol 2013;228:1658–64.

36. Alain T,MoritaM, Fonseca BD, Yanagiya A, SiddiquiN, BhatM, et al. eIF4E/4E-BP ratio predicts the efficacy of mTOR targeted therapies. Cancer Res2012;72:6468–76.

37. Lee J, Kotliarova S, Kotilarov Y, Li A, Su Q, Donin NM, et al. Tumor stemcells derived from glioblastomas cultured in bFGF and EGF more closelymirror the phenotype and genotype of primary tumors than do serum-cultured cell lines. Cancer Cell 2006;9:391–403.

38. Galli R, Binda E,OrfanelloU,Cipelletti B, Gritti A,De Vitis S, et al. Isolationand characterization of tumorigenic, stem-like neural precursors fromhuman glioblastoma. Cancer Res 2004;64:7011–21.

39. Laks DR, Masterman-Smith M, Visnyei K, Angenieux B, Orozco NM, ForanI, et al. Neurosphere formation is an independent predictor of clinicaloutcome in malignant glioma. Stem Cells 2009;27:980–7.

40. McGirt MJ, Chaichana KL, Gathinji M, Atenello F, Than K, Ruiz AJ, et al.Persistent outpatient hyperglycemia is independently associated withdecreased survival after primary resection ofmalignant brain astrocytomas.Neurosurgery 2008;63:286–291.

41. Silvera SAN, Meera Jain2, Howe Geoffrey R., Miller Anthony B., Rohan TE.Dietary carbohydrates and breast cancer risk: A prospective study of theroles of overall glycemic index and glycemic load. Int J Cancer 2005;114:653–8.

42. Ho VW, Leung K, Hsu A, Luk B, Lai J, Shen SY, et al. A low carbohydrate,high protein diet slows tumor growth and prevents cancer initiation.Cancer Res 2011;71:4484–93.

43. Zhang XD, Deslandes E, Villedieu M, Poulain L, Duval M, Gauduchon P,et al. Effect of 2-Deoxy-D-glucose on various malignant cell lines in vitro.Anticancer Res 2006;26:3561–6.

44. Maurer GD, Brucker DP, Bahr O, Harter PN, Hattingen E, Walenta S, et al.Differential utilization of ketone bodies by neurons and glioma cell lines: arationale for ketogenic diet as experimental glioma therapy. BMC Cancer2011;11:315.

45. Chang HT, Olson LK, Schwartz KA. Ketolytic and glycolytic enzymaticexpression profiles in malignant gliomas: implication for ketogenic diettherapy. Nutr Metab 2013;10:47–53.

46. Skinner R, Trujillo A,MaX, Beierle EA. Ketone bodies inhibit the viability ofhuman neuroblastoma cells. J Pediatr Surg 2009;44:212–6.

47. Shimazu T, HirscheyMD, Newman J, HeW, Shirakawa K, LeMoanN, et al.Suppression of oxidative stress by b -hydroxybutyrate, an endogenoushistone deacetylase inhibitor. Science 2013;339:211–4.

Clin Cancer Res; 22(10) May 15, 2016 Clinical Cancer Research2494

Martuscello et al.

Research. on September 18, 2020. © 2016 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 2, 2015; DOI: 10.1158/1078-0432.CCR-15-0916

Page 14: A Supplemented High-Fat Low-Carbohydrate Diet for the ... · Purpose:Dysregulated energetics coupled with uncontrolled proliferation has become a hallmark of cancer, leading to increased

48. Bose P, Yun Dai, Grant S. Histone deacetylase inhibitor (HDACI) mechan-isms of action: Emerging insights. Pharmacol Ther 2014;143:323–36.

49. Wu G Y, Thompson JR. The effect of ketone bodies on alanine andglutamine metabolism in isolated skeletal muscle from the fasted chick.Biochem J 1988;255:139–44.

50. Newman JC, Verdin E. Ketone bodies as signaling metabolites. TrendsEndocrinol Metab 2014;25:42–52.

51. Bonuccelli G, Tsirigos A, Whitaker-Menezes D, Pavildes S, Pestell RG,Chiavarina B, et al. Ketones and lactate "fuel" tumor growth andmetastasis:Evidence that epithelial cancer cells use oxidative mitochondrial metab-olism. Cell Cycle 2010;9:3506–14.

52. Cairns RA, Isaac S. Harris, Mak TW. Regulation of cancer cell metabolism.Nat Rev 2011;11:85–95.

53. OttoC,KaemmererU, Illert B,MuehlingB, PfetzerN,Wittig R, et al. Growthof 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;8:122–34.

54. Hao G-W, Chen Y-S, He D-M, Wang H-Y, Wu G-H, Zhang B. Growth ofhuman colon cancer cells in nudemice is delayed by ketogenic diet with orwithout omega-3 fatty acids and medium-chain triglycerides. Asian PacJ Cancer Prev 2015;16:2061–8.

55. Tisdale MJ, Brennan RA. A comparison of long-chain triglycerides andmedium-chain triglycerides on weight loss and tumour size in a cachexiamodel. Br J Cancer 1988;58:580–3.

56. Mcdaniel SS, RensingNR, Thio LL, YamadaKA,WongM.The ketogenic dietinhibits the mammalian target of rapamycin (mTOR) pathway. Epilepsia2011;52:e7–11.

57. Danial NN, Hartman AL, Stafstrom CE, Thio LL. How does the keto-genic diet work?: Four potential mechanisms. J Child Neurol 2013;0:1–7.

www.aacrjournals.org Clin Cancer Res; 22(10) May 15, 2016 2495

Supp. High-fat Low-Carbohydrate Diet for Glioblastoma

Research. on September 18, 2020. © 2016 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 2, 2015; DOI: 10.1158/1078-0432.CCR-15-0916

Page 15: A Supplemented High-Fat Low-Carbohydrate Diet for the ... · Purpose:Dysregulated energetics coupled with uncontrolled proliferation has become a hallmark of cancer, leading to increased

2016;22:2482-2495. Published OnlineFirst December 2, 2015.Clin Cancer Res   Regina T. Martuscello, Vinata Vedam-Mai, David J. McCarthy, et al.   of GlioblastomaA Supplemented High-Fat Low-Carbohydrate Diet for the Treatment

  Updated version

  10.1158/1078-0432.CCR-15-0916doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://clincancerres.aacrjournals.org/content/suppl/2015/12/02/1078-0432.CCR-15-0916.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://clincancerres.aacrjournals.org/content/22/10/2482.full#ref-list-1

This article cites 57 articles, 9 of which you can access for free at:

  Citing articles

  http://clincancerres.aacrjournals.org/content/22/10/2482.full#related-urls

This article has been cited by 6 HighWire-hosted articles. Access the articles at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected]

To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://clincancerres.aacrjournals.org/content/22/10/2482To request permission to re-use all or part of this article, use this link

Research. on September 18, 2020. © 2016 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 2, 2015; DOI: 10.1158/1078-0432.CCR-15-0916


Recommended