ArticlePDF AvailableLiterature Review

Effects of the Ketogenic Diet in the Treatment of Gliomas: A Systematic Review

MDPI
Nutrients
Authors:

Abstract and Figures

The ketogenic diet (KD) is a restrictive therapeutic diet, distinguished by being hyperlipidic, normoproteic, and hypoglucidic. This diet simulates biochemical changes related to fasting periods to achieve systemic ketosis. The metabolic particularities of glioma tumors motivated the rise in investigations and nutritional strategies, such as KD, to modulate the glycemic response as a treatment. This systematic review followed the PRISMA recommendations and was published in PROSPERO, with the identification CRD42021264173. The databases used were EMBASE, PubMed/Medline, Scopus, and Web of Science, and the studies were analyzed using the web-based application Rayyan. To analyze the risk of bias, Cochrane RevMan 5 software was used. For the analysis and treatment of statistical data, Microsoft® Excel® was used. A total of nine original articles were included. Data on survival, symptomology, and quality of life were collected. Mean overall survival was 15.9 months. Constipation and fatigue were the most reported symptoms. In 44.4% of the studies, an improvement in the quality of life was found. The KD is supported by most published studies as an effective therapy in the treatment of malignant gliomas due to its positive effects on patient survival. It was not possible to conclude the effectiveness of KD on quality of life.
This content is subject to copyright.


Citation: Sargaço, B.; Oliveira, P.A.;
Antunes, M.L.; Moreira, A.C. Effects
of the Ketogenic Diet in the
Treatment of Gliomas: A Systematic
Review. Nutrients 2022,14, 1007.
https://doi.org/10.3390/
nu14051007
Academic Editor: Keisuke Hagihara
Received: 9 February 2022
Accepted: 25 February 2022
Published: 27 February 2022
Publisher’s Note: MDPI stays neutral
with regard to jurisdictional claims in
published maps and institutional affil-
iations.
Copyright: © 2022 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
nutrients
Systematic Review
Effects of the Ketogenic Diet in the Treatment of Gliomas:
A Systematic Review
Beatriz Sargaço 1, 2, * , Patrícia Almeida Oliveira 2, Maria Luz Antunes 2,3 and Ana Catarina Moreira 2,4
1Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
2ESTeSL-Escola Superior de Tecnologia da Saúde de Lisboa, Instituto Politécnico de Lisboa,
1990-096 Lisboa, Portugal; patricia.aao@gmail.com (P.A.O.); mluz.antunes@estesl.ipl.pt (M.L.A.);
ana.moreira@estesl.ipl.pt (A.C.M.)
3APPsyCI–Applied Psychology Research Center Capabilities & Inclusion, ISPA–Instituto Universitário,
1149-041 Lisboa, Portugal
4H&TRC-Health & Technology Research Center, 1990-096 Lisboa, Portugal
*Correspondence: beatrizsargaconutri@gmail.com
Abstract:
The ketogenic diet (KD) is a restrictive therapeutic diet, distinguished by being hyperlipidic,
normoproteic, and hypoglucidic. This diet simulates biochemical changes related to fasting periods
to achieve systemic ketosis. The metabolic particularities of glioma tumors motivated the rise in
investigations and nutritional strategies, such as KD, to modulate the glycemic response as a treatment.
This systematic review followed the PRISMA recommendations and was published in PROSPERO,
with the identification CRD42021264173. The databases used were EMBASE, PubMed/Medline,
Scopus, and Web of Science, and the studies were analyzed using the web-based application Rayyan.
To analyze the risk of bias, Cochrane RevMan 5 software was used. For the analysis and treatment of
statistical data, Microsoft
®
Excel
®
was used. A total of nine original articles were included. Data on
survival, symptomology, and quality of life were collected. Mean overall survival was 15.9 months.
Constipation and fatigue were the most reported symptoms. In 44.4% of the studies, an improvement
in the quality of life was found. The KD is supported by most published studies as an effective
therapy in the treatment of malignant gliomas due to its positive effects on patient survival. It was
not possible to conclude the effectiveness of KD on quality of life.
Keywords: ketogenic diet; glioma; glioblastoma; survival
1. Introduction
The ketogenic diet (KD) is a restrictive therapeutic diet distinguished by being high-fat,
normoproteic, and hypoglucidic, and is considered a non-pharmacological therapeutic
method [
1
3
]. There are various types of KD according to the proportion of macronutrients.
The four major types of KD are the Classic Ketogenic Diet (CKD), the Ketogenic Diet
with Medium-Chain Triglycerides (MCT), the Modified Atkins Diet (MAD), and the Low
Glycemic Index Diet (LGID). The classical type is classified by a ratio of 3:1 to 4:1, that is, 3
to 4 grams of lipids for every 1 gram of carbohydrate and protein [
4
,
5
]. The utilization of
MCT as an energy source is due to the greater production of ketone bodies compared to
long-chain fatty acids.
Generally, the KD provides a high amount of lipids (60–90% of total energy value
(TEV)), a low carbohydrate content (4–19% TEV), and is adequate in protein, minerals, and
vitamins [6,7].
All of these diets have lipid restrictions, but in the CKD, the restriction is more severe.
Metabolically, carbohydrates break down into glucose. However, in the absence of this
energy source, the liver converts fat reserves into fatty acids and ketone bodies, allowing
the utilization of these as alternative sources of energy. Ketone bodies, such as acetone,
Nutrients 2022,14, 1007. https://doi.org/10.3390/nu14051007 https://www.mdpi.com/journal/nutrients
Nutrients 2022,14, 1007 2 of 17
acetoacetate, and ß-hydroxybutyrate, overcome the blood–brain barrier and replace glucose
usage [1,8,9].
KD simulates fasting periods by increasing ketones and lowering blood glucose, which
leads to increased oxidation of fatty acids and production of acetyl-CoA [10,11].
When acetyl-CoA production exceeds the capacity that can be used on the tricarboxylic acid
cycle, there is an increase in the production of ketone bodies, particularly
β
-hydroxybutyrate
and acetoacetate, that will be used as energy sources in the brain [1,8,11,12].
Gliomas are primary, heterogeneous, invasive, and aggressive malignancies, which
encompass most central nervous system tumors [
1
,
5
,
13
]. They originate from glial cells or
stem cells, and after neoplastic transformation, acquire glial cell characteristics [14].
The survival prognosis for patients with malignant gliomas is around 12 to 15 months,
with a survival rate of 5 years below 5% [
13
,
15
17
]. Thus, it is of high importance to develop
new therapeutic strategies for these patients, especially those that can improve quality of
life and/or survival [18,19].
The most frequent types of gliomas are glioblastomas, around 60–70% of all diagnosed
cases, followed by anaplastic astrocytomas with 10–15% of cases and, finally, anaplastic
oligodendrogliomas and anaplastic oligoastrocytomas with 10% of cases. The remainder
includes gliomas such as anaplastic ependymomas and anaplastic gangliogliomas [20,21].
The metabolic particularities of cancer have motivated increased research on nutri-
tional strategies to modulate the glycemic response during treatment. These nutritional
strategies include the use of diets with high lipidic content and low carbohydrate content
(such as KD), energy-restricted diets, or intermittent fasting [
22
]. The purpose of these diets
is to induce a state of systemic ketosis to compensate for the decrease in glucose, originated
from the lack of substrate [19].
In tumors, there are changes in the metabolism of cancer cells. These changes can be
explained by the "Warburg effect", also known as aerobic glycolysis [
18
]. Otto Warburg,
in 1924, observed that tumors had a metabolic phenotype with high rates of aerobic
glycolysis [3].
Tumor cells depend on mitochondrial oxidative phosphorylation to generate adenosine
triphosphate (ATP). Thus, when exposed to hypoxia, they switch to the less favorable
anaerobic pathway of glycolysis. However, there are cancer cells that survive and proliferate
generating ATP via glycolysis instead of oxidative phosphorylation, even in the presence
of oxygen [1,8,15,18].
Cancer cells differ from other cells in their inability to use ketones as metabolic fuel.
Tumor cells are very glucose-dependent for their growth and survival, while the remaining
cells have the flexibility to alter their energy source from glucose to ketone bodies. With
KD, tumor cells would no longer have an energy source for their growth [1,8,15,18].
By reducing glucose availability and providing ketone bodies as an alternative energy
source, KD can offer a therapeutic approach directing the Warburg effect on glycolytic
tumors such as malignant gliomas. Nutritional strategies aimed at glycemic modula-
tion to explore tumor cells’ dependence on glucose have not yet been fully studied and
investigated, so the existing clinical data are limited [19,22].
There have been several studies with the inclusion of this type of diet in the therapeutic
intervention of diagnosed gliomas. Although systematic reviews addressing the impact
of KD on the survival of patients with gliomas have been published in the past, this
area of research has been intensively studied further, with some relevant original studies
published since the latest review. The systematic analysis of the effect of this nutritional
intervention covering the different types of diets, side effects, and quality of life is not yet
well established. KD implementation raises the risk of many side effects, so it is essential to
analyze this strand and all relevant studies published to date in a systematic review.
2. Materials and Methods
This systematic review followed the recommendations of PRISMA (Preferred Report-
ing Items for Systematic Reviews and Metanalyses), which is intended for systematic
Nutrients 2022,14, 1007 3 of 17
reviews and meta-analyses of clinical intervention studies [
23
]. This systematic review was
published on the platform PROSPERO, the International Prospective Register of Systematic
Reviews [24], on 17 July 2021 with the registration number CRD42021264173.
The used databases were EMBASE, PubMed/Medline, Scopus, and Web of Science,
with the following keyword strategies:
[(glioma) OR (glial cell tumor) OR (mixed glioma) OR (malignant glioma) OR (glioblas-
toma) OR (astrocytoma, grade IV) OR (glioblastoma multiforme) OR (giant cell
glioblastoma) OR (high-grade glioma) OR (neoplastic stem cell) OR (cancer stem
cells) OR (Warburg effect, oncologic) OR (astrocytoma) OR (brain tumor) OR (brain
neoplasms) OR (glial cell) OR (glial precursors) OR (astrocytic tumor) OR (oligoastro-
cytic tumor) OR (neuro-glial tumor)] AND [(ketogenic diet) OR (low carbohydrate
diet) OR (glucose) OR (glycolysis) OR (ketone bodies) OR (low carbohydrate) OR (diet
therapy) OR (ketosis) OR (caloric restriction) OR (therapeutic ketosis) OR (low carb
diet) OR (metabolic therapy) OR (adjuvant therapy) OR (energetic restriction) OR
(energy restriction) OR (high-fat diet) OR (low carbohydrate high-fat diet) OR (keto-
induction) OR (ketotherapy) OR (glucose restriction) OR (carbohydrate-restriction)
OR (low carb) OR (hyper lipidic diet) OR (high-fat diet) OR (dietary intervention)].
The research included original articles published between January 2005 and May 2021
that included KD as a therapeutic intervention for the treatment of gliomas. After remov-
ing duplicate papers, the article selection was carried out according to titles, abstracts,
and full texts. Case-control studies, retrospective and prospective cohort studies, and
randomized clinical trials were included. The web-based application Rayyan, a software
for systematic reviews and meta-analyses to facilitate initial screening through titles and
abstracts, was used to analyze the identified studies [
25
]. This analysis for the identification
of relevant publications was practiced by two reviewers who examined the articles inde-
pendently for their inclusion, not knowing each other’s decisions. All divergences were
resolved by consensus, and the divergences that did not reach consensus were resolved by
a third reviewer.
For the analysis of the risk of bias, software designed for the development of systematic
reviews and meta-analyses, Cochrane RevMan 5, was used. All the studies obtained were
evaluated in depth by two reviewers regarding the risk of various biases to evaluate the
quality of evidence. The risk of bias was classified as high risk, low risk, uncertain, or
unapplicable risk [26].
Information was collected on the type of study (controlled or uncontrolled), the
study design (prospective or retrospective), number of patients, age of patients, type of
investigated glioma, stage of the disease, type of interventions performed, follow-up time,
and type and duration of KD.
An initial survey was carried out on 24 April 2021 and repeated on 7 June 2021 to
verify that all recent studies were covered, including the latest ones.
Microsoft
®
Excel
®
for Mac, version 16.41 (Microsoft Company), was used for the
analysis and processing of statistical data.
2.1. Inclusion and Exclusion Criteria
For this systematic review, inclusion criteria were considered:
Original studies, published between 1 January 2005 and 30 May 2021;
Clinical studies;
Studies evaluating patients with malignant gliomas;
Studies using the KD as an intervention/treatment for malignant gliomas;
Studies published in English or Portuguese;
Studies including patients of all ages, genders, and ethnicities in any country.
For the exclusion criteria, we considered:
Articles such as comments, letters to the editor, reviews, conference editorials, opinion
articles, or case reports;
Nutrients 2022,14, 1007 4 of 17
Articles associating KD with non-conventional therapies (e.g., intermittent fasting or
perillyl alcohol);
Studies that did not include results;
Animal studies.
3. Results
The procedure for the study selection in this systematic review is presented in Figure 1.
Nutrients 2022, 13, x FOR PEER REVIEW 4 of 17
Original studies, published between 1 January 2005 and 30 May 2021;
Clinical studies;
Studies evaluating patients with malignant gliomas;
Studies using the KD as an intervention/treatment for malignant gliomas;
Studies published in English or Portuguese;
Studies including patients of all ages, genders, and ethnicities in any country.
For the exclusion criteria, we considered:
Articles such as comments, letters to the editor, reviews, conference editorials,
opinion articles, or case reports;
Articles associating KD with non-conventional therapies (e.g., intermittent fast-
ing or perillyl alcohol);
Studies that did not include results;
Animal studies.
3. Results
The procedure for the study selection in this systematic review is presented in Figure
1.
Figure 1. PRISMA flow chart—study selection procedure.
Figure 1. PRISMA flow chart—study selection procedure.
The bibliographic research in PubMed/Medline, Scopus, and Web of Science databases
resulted in 6071 results. This research was supplemented by research in previously pub-
lished systematic reviews, which identified two additional studies. After excluding non-
relevant studies, a total of nine original articles were included.
The analysis of the included studies regarding the type of study, study design, number
of patients, age of patients, type of investigated glioma, stage of the disease, type of
interventions performed, follow-up time, and type and duration of the KD is presented in
Table 1.
Table 2presents the detailed data regarding patient survival, symptoms, and quality
of life.
Nutrients 2022,14, 1007 5 of 17
3.1. General Characteristics of Studies
The studies included in this review were published between 2014 and 2021, and more
recent data were included in this systematic review in comparison with previous reviews.
Of the nine included studies, six (66.7%) were prospective studies and two (22.2%)
had a control group. In most studies, it was found that the number of included patients
was low, with an average of 8.3 (3 to 17) patients per study.
The age of patients included in the studies ranged from 5.3 years old to 72 years
old. Two studies were conducted in children between 2.5 and 15 years old, and 5.3 and
15.5 year
s old. The remaining studies were performed on adults aged between 45 and
72 year
s old. The overall mean age of patients with gliomas included in the analyzed
studies was 42.1 years old, with a standard deviation of 19.9 years old.
The time of dietary intervention ranged from three weeks to 26 months. The follow-up
period ranged from 12 weeks to 26 months.
3.2. Characteristics of Gliomas
Concerning the type of investigated glioma, the studies observed elevated stage
gliomas such as glioblastomas in 66.7% of the studies, astrocytomas in 22.2% of the studies,
intrinsic pontine gliomas in 22.2% of the studies, and glioblastomas multiforme in 11.1%
of the studies. Grade II (22.2%) and grade III (33.3%) gliomas were included, and patients
with grade IV glioma were present in all studies.
Regarding the treatment of gliomas, in these studies, all patients underwent joint
radiotherapy with temozolomide. However, these were not the only interventions applied.
Drugs such as temozolomide, gemcitabine, prednisolone, bevacizumab, and lomustine
were applied individually in four different studies (33.3%): temozolomide in the studies
by van der Louw et al., 2019b [
15
] and Perez et al. [
27
]; bevacizumab and lomustine in
the study by Rieger et al. [
8
]; lomustine in the study by Martin-McGill et al., 2018 [
17
];
and gemcitabine and prednisolone in the study by van der Louw 2019a [
10
]. In one study,
patients received chemotherapy and concomitant radiotherapy as therapeutic interventions
since the initial treatment with radiotherapy had no effect [28].
3.3. Features of the Implemented Diet
In the analyzed studies, we verified the implementation of different types of KD.
In the study by Rieger et al., KD restricted carbohydrate intake to 60 g per day,
including intake of 500 mL of highly fermented yogurt drinks and two vegetable oils. No
energy restriction was applied [8].
Martin-McGill et al., 2018 applied a Modified Ketogenic Diet (MKD) comprising 70%
dietary lipids, while carbohydrates were limited to 20 g per day, corresponding to 3–5% of
TEV. The protein content had no restriction [17].
In the study by van der Louw et al., 2019b, KD included two phases. During the initial
eight weeks, associated with radiotherapy and temozolomide, KD was exclusively liquid
with a ratio of 4:1. Over the next six weeks, KD transitioned to solid foods at a ratio of
1.5–2.0:1, with MCT [15].
Champ et al. restricted carbohydrate intake to <50 g per day, corresponding to 8% of
TEV. Lipids accounted for 77% of the energy value and proteins 15% [16].
In the study by van der Louw et al. 2019a, patients started KD with a commercial
liquid enteral formula at a ratio of 4:1 for a maximum of two weeks. When ketone levels
reached 3 mmol/L, the KD was changed to a ratio of 1.5:1–2.0:11. During the study, MCTs
were administered with KD [10].
In the study by Klein et al., the authors applied a KD in a ratio of 4:1 in two different
groups. In group 1 included patients with recent glioblastoma; in group 2, patients with
recurrent glioblastoma were included. The diet included 1600 kcal per day, with a total of
10 g of carbohydrates [29].
Nutrients 2022,14, 1007 6 of 17
Perez et al. grouped patients undergoing various types of KD, including KD with
MCT, CKD, and MAD. However, this study did not specify the proportions or amounts of
macronutrients applied [27].
Martin-McGill et al. 2020 divided patients into two groups with the aim of imple-
menting two different KDs: group 1 corresponded to a KD with MCT, and group 2 to an
MKD. However, as in the study by Perez et al., no proportion or amount of administered
macronutrients were reported [30].
In the study by Panhans et al., a 3:1 ratio KD was applied. The intake of carbohydrates
was limited to 20 g per day [28].
Although the analyzed studies implement different types of KD, we can verify that
the studies by Martin-McGill et al., 2018, Klein et al., and Panhans et al. applied more
restrictive diets, having a maximum carbohydrate intake of 20 g per day. On the other
hand, the studies by Rieger et al. and Champ et al. implemented more liberal KDs with
daily carbohydrate intakes of 60 g and 50 g, respectively.
3.4. Overall Patient Survival
Patient survival varied from study to study. The study by Martin-McGill 2018 et al.
made no reference to patient survival.
The mean of maximum overall survival was 25.4 months in the study by Klein et al.,
while the lower overall survival corresponded to the study by Rieger et al., with a median
of 32 weeks (8 months) of survival [8,29].
Rieger et al. implemented KD for 16 weeks, where ketosis was reached and kept
stable in most patients. There was stabilization of the disease at six weeks of diet, with
this stabilization being balanced in an average duration of 12 weeks. The median overall
survival after starting the diet was 32 weeks (
8 months), ranging between 6 weeks
(1.5 months) and 86 weeks (21.5 months) [8].
Overall survival in the study by Champ et al. had an average of 14 months, and the
recurrence of the disease was, on average, 10.3 months. KD was implemented throughout
the follow-up period (14 months) [16].
In the study by van der Louw et al. 2019a, the KD implementation lasted three months.
In this study, diffuse intrinsic pontine gliomas that have a median overall survival of 9 to
11 months were evaluated [
10
]. The overall survival times observed in this study were 6.4,
16.5, and 18.7 months for each of the participants. The 6.4-month survival was explained
by a severe decrease in consciousness after a generalized tonic–clonic crisis in one of the
patients, who had only completed three weeks of the dietary intervention. Survival after
starting nutritional therapy was 6.5 months for the remaining cases [10].
In the study by van der Louw et al. 2019b, overall survival averaged 12.8 months,
with two patients achieving survival times of 17.7 and 19 months. Ketogenic therapy was
performed for 14 weeks, and patients reached ketosis after an average of 4.5 days [15].
The study by Klein et al. was divided into two groups. Both groups received KD as
therapy, with group 1 including patients with recent glioblastoma and group 2 including
patients with recurrent glioblastoma. KD was implemented for six months. The mean
survival from the onset of KD was 20 months for group 1 and 12.8 months for group 2.
However, in terms of overall survival, group 1 had an average of 21.8 months and group 2
had an average of 25.4 months [29].
Nutrients 2022,14, 1007 7 of 17
Table 1. Overview of studies regarding their design, subjects, type of investigated glioma, and therapeutic interventions.
Study Type of Study Study Design NAge of Patients Type of Glioma Disease Stage Interventions KD Duration Follow-up Time Type of KD
Rieger 2014 [8] Controlled Prospective 17 Med 57 years
(30–72) Glioblastoma Grade IV
RT +
Temozolomide;
Bevacizumab and
Lomustine
3 to 16 weeks 16 weeks
Max. 60 g carbs/day;
highly fermented yogurt
drinks (500 ml per day)
and two different
vegetable oils (base oil
and addition oil)
Champ 2014 [16] Not controlled Retrospective 6 Med 54 years
(34–62) Glioblastoma Grade III–IV RT +
Temozolomide 14 months 14 months KD 50 g carbs/day
Martin-McGill 2018 [17] Controlled Prospective 6 Med 46 years
(34–49)
Glioblastoma
Anaplastic
astrocytoma Grade II–IV RT +
Temozolomide;
Lomustine 12 weeks 12 weeks Max. 20 g carbs/day,
70% lipids
van der Louw 2019a [10] Not controlled Prospective 3 Med 11.6 years
(5.3–15.5)
Diffuse intrinsic
pontine glioma Grade IV
RT +
Temozolomide;
Gemcitabine;
Prednisolone;
Temozolomide
3 months 3 months Liquid KD: 4:1 (+MCT)
van der Louw 2019b [15] Not controlled Prospective 9 Med 53.8 years
(33.5–65.5) Glioblastoma Grade IV RT +
Temozolomide 14 weeks 14 weeks
Liquid KD 4:1 (8 weeks):
11 g carbs
solid KD + MCT 1.5-2:1
(6 weeks): 57 g carbs
Klein 2020 [29] Not controlled Prospective 5 Med 49.8 years
(40–64) Glioblastoma Grade IV RT +
Temozolomide 6 to 26 months 26 months KD 4:1; 10 g carbs/day
Martin-McGill 2020 [30] Controlled Prospective 12 Med 57 years
(44–66) Glioblastoma Grade IV RT +
Temozolomide
38 days to
12 months 12 months KD with
MCT/Modified KD
Panhans 2020 [28] Not controlled Retrospective 12 Med 45 years
(32–62)
Glioblastoma
multiforme,
astrocytoma, oligo-
dendroglioma
Grade II–IV Chemotherapy +
RT; RT; RT +
Temozolomide 120 days 120 days KD 3:1; 20 g carbs/day
Perez 2021 [27] Not controlled Retrospective 5 Med 4.4 years
(2.5–15)
Diffuse intrinsic
pontine glioma Grade IV
RT +
Temozolomide;
Chemotherapy
HIT-SKK;
Temozolomide
6.5 months (0.25 to
2 years) 2 years Classic KD/KD with
MCT/Modified
Atkins Diet
RT, radiotherapy; Med, median; MCT, medium-chain triglycerides; carbs, carbohydrates; N, number of patients; KD, ketogenic diet; Max., maximum; HIT-SKK, Therapieprotokoll für
Säuglinge und Kleinkinder mit Hirntumoren (Therapy protocol for infants and young children with brain tumors).
Nutrients 2022,14, 1007 8 of 17
Table 2. Overview of study results on survival, symptoms, and quality of life.
Study Patient Survival Symptomatology Associated with Dietary
Intervention Quality of Life
Rieger 2014 [8] Med 32 weeks (between 6 and 86 weeks). Weight loss, diarrhea, constipation, hunger Decreased quality of life
Champ 2014 [16] Med 14 months Constipation, asthenia, weight loss,
nephrolithiasis, hypoglycemia Not available
Martin-McGill 2018 [17] Not available Constipation Improved quality of life
van der Louw 2019a [10] Between 16.5 and 18.7 months Hypoglycemia, hyperketosis, vomiting,
refusal to eat, asthenia, constipation Decreased quality of life
van der Louw 2019b [15] Between 9.8 and 19.0 months (Med 12.8 months)
Constipation, nausea/vomiting,
hypercholesterolemia, hypoglycemia,
diarrhea, low carnitine concentration
Decreased quality of life
Klein 2020 [29]Group 1: ¯
x=21.9 months (between 11 and 29.2 months)
Group 2: ¯
x=25.4 months (between 13.9 and 38.7 months)
Weight loss, hunger, nausea, dizziness,
asthenia, constipation Improved quality of life
Martin-McGill 2020 [30] Med 67.3 weeks Hypokalemia, hypocalcemia, hypernatremia,
hyperkalemia, constipation Improved quality of life
Panhans 2020 [28] Between 9.8 and 19.0 months Asthenia, weight loss, nausea, vomiting,
headache, decreased appetite Improved quality of life
Perez 2021 [27] Med 18.7 months Hypoglycemia, constipation, hyperketosis,
vomiting, asthenia, hyperuricemia Not available
Med, median; ¯
x, mean.
Nutrients 2022,14, 1007 9 of 17
Martin-McGill et al., 2020 divided the patients into two groups, distinguishing the
implemented KDs. Group 1 corresponded to a KD with MCT and group 2 to an MKD. In
both groups, ketosis was reached within the first six weeks. Overall survival had a median
of 67.3 weeks (16.8 months) [30].
Panhans et al. applied KD for at least 120 days. Ketosis was achieved in most patients
within the first week of therapy. Overall survival ranged from 9.8 to 19 months [28].
The study by Perez et al. included patients who had implemented ketogenic therapy
for a period longer than three months. The estimated overall survival was 18.7 months.
There were also two patients who achieved survival times of 22 and 30 months [27].
The study by Martin-McGill et al., 2018 did not report patient survival [
17
,
31
]. How-
ever, the KD was implemented for 12 weeks, and three patients continued this therapy for
more than 360 days [17], which is a possible assumption regarding survival in this study.
3.5. Associated Symptomatology
The symptoms associated with KD intervention detailed in the studies included
75 patients and are shown in Figure 1.
In Figure 2, we can see that constipation was the most reported symptom in the
analyzed studies (88.9%). This was followed by asthenia, which was reported in 55.6% of
the studies.
Nutrients 2021, 13, x FOR PEER REVIEW 5 of 17
Nutrients 2022, 13, x. https://doi.org/10.3390/xxxxx www.mdpi.com/journal/nutrients
Martin-McGill et al. 2020 divided the patients into two groups, distinguishing the
implemented KDs. Group 1 corresponded to a KD with MCT and group 2 to an MKD. In
both groups, ketosis was reached within the first six weeks. Overall survival had a median
of 67.3 weeks ( 16.8 months) [30].
Panhans et al. applied KD for at least 120 days. Ketosis was achieved in most patients
within the first week of therapy. Overall survival ranged from 9.8 to 19 months [28].
The study by Perez et al. included patients who had implemented ketogenic therapy
for a period longer than three months. The estimated overall survival was 18.7 months.
There were also two patients who achieved survival times of 22 and 30 months [27].
The study by Martin-McGill et al. 2018 did not report patient survival [17,31]. How-
ever, the KD was implemented for 12 weeks, and three patients continued this therapy for
more than 360 days [17], which is a possible assumption regarding survival in this study.
3.5. Associated Symptomatology
The symptoms associated with KD intervention detailed in the studies included 75
patients and are shown in Figure 1.
Figure 2. Symptomatology associated with KD therapy in glioma patients.
In Figure 2, we can see that constipation was the most reported symptom in the ana-
lyzed studies (88.9%). This was followed by asthenia, which was reported in 55.6% of the
studies.
In 44.4% of the studies, weight loss, vomiting, or hypoglycemia were reported as as-
sociated symptoms, whereas in 33.3% of the studies, nausea was reported.
Patients reported diarrhea, hunger, or hyperketosis as associated symptoms in 22.2%
of the analyzed studies, and only 11.1% of the studies reported dizziness, anorexia, food
refusal, hypernatremia, hypokalemia, hypocalcemia, low carnitine concentration, head-
ache, hyperuricemia, or hypercholesterolemia.
Important symptoms associated with KD include hypoglycemia, weight loss, vomit-
ing, diarrhea, and nausea. In this review, hypoglycemia was reported in the studies by
Champ et al., van der Louw et al. 2019a and 2019b, and Perez et al. Hypoglycemia, re-
ported in the study by Champ et al., was asymptomatic [16]. Weight loss was reported in
studies by Rieger et al. [8], Champ et al. [16], Klein et al. [29], and Panhans et al. [28].
Vomiting was reported in the two studies by van der Louw et al. [10,15], in the study by
Panhans et al. [28], and in that of Perez et al. [27]. Nausea was reported in the studies by
88.9%
55.6%
44.4%
44.4%
44.4%
33.3%
22.2%
22.2%
22.2%
11.1%
11.1%
11.1%
11.1%
11.1%
11.1%
11.1%
11.1%
11.1%
11.1%
11.1%
Constipation
Asthenia
Weight loss
Vomiting
Hypoglycemia
Nausea
Diarrhea
Hunger
Hyperketosi
Hypercholesterolemia
Hyperuricemia
Headache
Low carnitine concentration
Hypokalemia
Hypernatremia
Hypocalcemia
Anorexia
Dizziness
Nephrolithiasis
Refusal to feed
Symptomatology
Figure 2. Symptomatology associated with KD therapy in glioma patients.
In 44.4% of the studies, weight loss, vomiting, or hypoglycemia were reported as
associated symptoms, whereas in 33.3% of the studies, nausea was reported.
Patients reported diarrhea, hunger, or hyperketosis as associated symptoms in 22.2%
of the analyzed studies, and only 11.1% of the studies reported dizziness, anorexia, food
refusal, hypernatremia, hypokalemia, hypocalcemia, low carnitine concentration, headache,
hyperuricemia, or hypercholesterolemia.
Important symptoms associated with KD include hypoglycemia, weight loss, vomiting,
diarrhea, and nausea. In this review, hypoglycemia was reported in the studies by Champ
et al., van der Louw et al., 2019a and 2019b, and Perez et al. Hypoglycemia, reported in
the study by Champ et al., was asymptomatic [
16
]. Weight loss was reported in studies
by Rieger et al. [
8
], Champ et al. [
16
], Klein et al. [
29
], and Panhans et al. [
28
]. Vomiting
was reported in the two studies by van der Louw et al. [
10
,
15
], in the study by Panhans
et al. [
28
], and in that of Perez et al. [
27
]. Nausea was reported in the studies by van der
Louw et al., 2019b [
15
], Klein et al. [
29
], and Panhans et al. [
28
]. Diarrhea was referenced in
Nutrients 2022,14, 1007 10 of 17
the studies by Rieger et al. [
8
] and van der Louw et al., 2019b [
15
]. The authors referred to
weight loss as a minimal side effect [8,16,17].
It should be noted that in the study by van der Louw et al., 2019b, patients with
dexamethasone were excluded [
15
], while in the studies by Champ et al., Rieger et al.,
Martin-McGill et al., 2018, Panhans et al., Klein et al., and Martin-McGill et al., this exclusion
was not made, which could influence the symptomatology and the interpretation of blood
glucose values [8,16,17,2830].
Only one study reported the occurrence of hypercholesterolemia [
10
]. The remaining
studies did not report changes in the lipid profile.
3.6. Quality of Life
Quality of life is a parameter that must be evaluated with appropriate objective or
subjective measures, with the implementation of a validated questionnaire, for example.
Only three studies applied quality of life perception questionnaires [
8
,
15
,
30
]. The remaining
studies presented subjective results.
The perception of quality of life felt by patients with the implementation of ketogenic
therapy is shown in Figure 3.
Nutrients 2022, 13, x FOR PEER REVIEW 6 of 17
van der Louw et al. 2019b [15], Klein et al. [29], and Panhans et al. [28]. Diarrhea was
referenced in the studies by Rieger et al. [8] and van der Louw et al. 2019b [15]. The authors
referred to weight loss as a minimal side effect [8,16,17].
It should be noted that in the study by van der Louw et al. 2019b, patients with dex-
amethasone were excluded [15], while in the studies by Champ et al., Rieger et al., Martin-
McGill et al. 2018, Panhans et al., Klein et al., and Martin-McGill et al., this exclusion was
not made, which could influence the symptomatology and the interpretation of blood glu-
cose values [8,16,17,2830].
Only one study reported the occurrence of hypercholesterolemia [10]. The remaining
studies did not report changes in the lipid profile.
3.6. Quality of Life
Quality of life is a parameter that must be evaluated with appropriate objective or
subjective measures, with the implementation of a validated questionnaire, for example.
Only three studies applied quality of life perception questionnaires [8,15,30]. The remain-
ing studies presented subjective results.
The perception of quality of life felt by patients with the implementation of ketogenic
therapy is shown in Figure 3.
Figure 3. Perception of quality of life in glioma patients undergoing KD.
As previously stated, not all studies analyzed the quality of life of patients undergo-
ing treatment—22.2% did not report any results on quality of life, since they were not
indicators studied by the authors themselves.
In 44.4% of the studies, there was an improvement in the quality of life with the im-
plementation of KD, regardless of the symptoms felt.
In 33.3% of the studies, there was a decrease in the quality of life. In two of these
studies (Rieger et al. and van der Louw et al. 2019b), the authors estimated that this de-
crease was related to the symptoms experienced throughout the studies and to tumor pro-
gression [8,15].
Martin-McGill et al. 2020 reported that patients who withdrew from KD therapy jus-
tified it with a decrease in quality of life in comparison to the beginning of treatment.
However, patients who remained on therapy reported an improvement in quality of life,
as they had a sense of control during tumor treatment [30].
Studies that did not objectively assess the quality of life assumed a good quality of
life or an improvement in quality of life. Martin-McGill et al. 2018 reported a good quality
of life for patients, with an improvement in symptoms [17].
Figure 3. Perception of quality of life in glioma patients undergoing KD.
As previously stated, not all studies analyzed the quality of life of patients undergoing
treatment—22.2% did not report any results on quality of life, since they were not indicators
studied by the authors themselves.
In 44.4% of the studies, there was an improvement in the quality of life with the
implementation of KD, regardless of the symptoms felt.
In 33.3% of the studies, there was a decrease in the quality of life. In two of these
studies (Rieger et al. and van der Louw et al., 2019b), the authors estimated that this
decrease was related to the symptoms experienced throughout the studies and to tumor
progression [8,15].
Martin-McGill et al., 2020 reported that patients who withdrew from KD therapy
justified it with a decrease in quality of life in comparison to the beginning of treatment.
However, patients who remained on therapy reported an improvement in quality of life, as
they had a sense of control during tumor treatment [30].
Studies that did not objectively assess the quality of life assumed a good quality of life
or an improvement in quality of life. Martin-McGill et al., 2018 reported a good quality of
life for patients, with an improvement in symptoms [17].
The study by Panhans et al. mentioned that a qualitative assessment of the quality of
life was carried out, in which there was an improvement in energy, mood, neurocognitive
function, general well-being, and symptoms [28].
Nutrients 2022,14, 1007 11 of 17
Klein et al. did not assess the quality of life. Nonetheless, they referred to a patient
who improved his quality of life. It should be noted, however, that in this study, the meals
were prepared by a company and provided free of charge to patients [29].
In the study by van der Louw et al., 2019a, it was mentioned that KD was not com-
patible with a better quality of life for patients [
10
], which led to the understanding that
with the implementation of KD, there was a decrease in the quality of life. In this study,
the children were in palliative care, and hospitalization of two children was necessary for
the management of epileptic seizures and implementation of enteral nutritional support
through a nasogastric tube due to swallowing difficulties. These data were not related
to the implementation of KD, however, with direct influence in the perceived quality of
life [10].
The studies by Champ et al. and Perez et al. did not make any reference to the patients’
perception of quality of life during or after treatment with implementation of KD [
16
,
27
,
32
].
Even so, in the study by Perez et al., it was mentioned that patients had an improvement in
symptoms [27].
3.7. Bias Risk Analysis
The results of the quality assessment of the clinical trials are shown in Figure 4. To
assess the risk of bias, Cochrane RevMan 5 software (Review Manager 5.4.1) [
26
] was used.
Nutrients 2022, 13, x FOR PEER REVIEW 8 of 17
Figure 4. Risk of bias summary—risk of bias analysis for each included study [8,10,1517,2730].
4. Discussion
Evidence for the use of KD in clinical practice is still very limited. There has been an
attempt to further investigate this topic, which implies new studies to be implemented in
the search for more reliable, promising, and relevant results.
The main objective of the present systematic review was to analyze and systematize
the clinical studies that tested KD in the context of glioma treatments, focusing the analy-
sis of the evidence on the potential therapeutic value of KD as a treatment option in patient
survival, and to further evaluate the potential effects of KD on associated symptoms and
quality of life of these patients. A total of nine original studies that reported at least one
of the study objectives were identified and analyzed.
It should be noted that, of the nine studies included, two were applied to children
between 2.5 and 15 years old and 5.3 and 15.5 years old. The remaining studies were ap-
plied to adults aged between 45 and 72 years old.
4.1. Overall Patient Survival
KD, being a therapy that reduces blood glucose levels and increases ketone body lev-
els, can be an effective therapy to increase the survival of patients with gliomas [32].
In the studies included in this review, we found that different results were described,
with survival values between 32 weeks (approximately eight months) and 25.4 months.
Survival is a factor that may vary mainly with age, disease stage, adherence to therapy,
Figure 4. Risk of bias summary—risk of bias analysis for each included study [8,10,1517,2730].
Nutrients 2022,14, 1007 12 of 17
The performance bias includes the knowledge of the interventions by the participants
and the team that is part of the study. Due to uncontrolled studies, there was no blinding,
which led to a high risk of performance bias.
The detection bias refers to the concealment of the investigators about the therapeutic
interventions in order not to influence the results. There was a high risk of detection bias
because the authors were not blinded to the diet therapy.
Attrition bias encompasses the tendency to attrition due to the quantity, nature, or
manipulation of incomplete result data. Data on patient survival (essential variable),
symptomatology, and quality of life (secondary variables) were evaluated.
A high risk of attrition bias due to incomplete assessment of the quality of life outcomes
was notable in most studies. Only the study by van der Louw et al. 2019b presented a
low risk of bias [
15
], being the only study that applied a quality of life questionnaire and
presented those results.
We verified a low risk of attrition bias regarding the nature of the data and the
evaluation of the results for survival in four studies [
8
,
10
,
15
,
30
]. In the remaining four
studies, there was a high risk of bias [
16
,
27
29
] since it was observed that survival was
reported by observation of patients or through qualitative data, so there was no application
of statistical tests.
Regarding symptoms, we found a low risk of attrition bias for most studies [
8
,
10
,
15
,
16
,
29
].
However, three studies showed a high risk of bias [
17
,
27
,
28
]: the study by Martin-McGill et al.
2018 and by Panhans et al., due to the reporting of symptoms (no questionnaire or structured
interview method was applied), and in the study by Perez et al., as results were presented for
only three patients, with incomplete data. The study by Martin-McGill et al., 2020 presented
an uncertain risk of bias [
30
] and reported that they evaluated ketosis, food acceptability, and
gastrointestinal adverse events and, later, also presented mineral deficits.
Reporting bias considers bias due to the description of selective results. The studies by
Champ et al., Klein et al., Martin-McGill et al., 2018, Martin-McGill et al., 2020, and Rieger
et al. were at high risk of bias [
8
,
16
,
17
,
29
,
30
]. The studies by van der Louw et al., 2019a and
2019b had a low risk of bias [10,15].
4. Discussion
Evidence for the use of KD in clinical practice is still very limited. There has been an
attempt to further investigate this topic, which implies new studies to be implemented in
the search for more reliable, promising, and relevant results.
The main objective of the present systematic review was to analyze and systematize
the clinical studies that tested KD in the context of glioma treatments, focusing the analysis
of the evidence on the potential therapeutic value of KD as a treatment option in patient
survival, and to further evaluate the potential effects of KD on associated symptoms and
quality of life of these patients. A total of nine original studies that reported at least one of
the study objectives were identified and analyzed.
It should be noted that, of the nine studies included, two were applied to children
between 2.5 and 15 years old and 5.3 and 15.5 years old. The remaining studies were
applied to adults aged between 45 and 72 years old.
4.1. Overall Patient Survival
KD, being a therapy that reduces blood glucose levels and increases ketone body
levels, can be an effective therapy to increase the survival of patients with gliomas [32].
In the studies included in this review, we found that different results were described,
with survival values between 32 weeks (approximately eight months) and 25.4 months.
Survival is a factor that may vary mainly with age, disease stage, adherence to therapy,
treatment efficacy, and tumor location. In this review, the mean overall survival was found
to be around 15.9 months.
The survival interval of patients with gliomas on common therapy is between 12 and
15 months [
13
]. With the implementation of KD, it was possible to verify that survival in the
Nutrients 2022,14, 1007 13 of 17
studies by Champ et al. and van der Louw et al. 2019b corresponded to this interval [
16
],
with no improvement in comparison to the survival time considered in the literature.
Patients included in the studies by van der Louw et al., 2019a, Klein et al., Martin-McGill
et al., 2020, and Perez et al. exceeded the average survival of those undergoing common
therapy [
10
,
27
,
30
]. The study by Rieger et al. was the only one in which the survival time
was lower than the average time of patients with gliomas undergoing common therapy [
8
].
According to the authors, the reasons for the low clinical activity may be due to the failure
to significantly reduce glucose through KD or to the hypothesis that tumor cells bypassed
the glucose reduction using ketone bodies [8].
The study that showed a longer survival was reported by Klein et al., where a KD
with a ratio of 4:1 was implemented, with 1600 kcal per day and a total of 10 g of carbohy-
drates [
29
]. On the other hand, patients from Rieger et al., who implemented a KD without
energy restriction but with restriction of carbohydrate intake to 60 g per day, had the lowest
survival [8].
In this analysis, the type of KD that presented the highest overall survival was CKD,
with a ratio of 4:1, verified in the studies by Klein et al. and van der Louw et al. 2019b.
Although the studies by Martin-McGill et al. and Perez et al. showed an increase in overall
survival, they applied different types of KD, with no description of the proportions of
macronutrients provided.
In most of the studies analyzed with the implementation of KD therapy, the overall
survival exceeded the prognosis of patients with the usual chemotherapy and/or radio-
therapy associated with temozolomide. A previously published review also mentions that
KD improved the survival of most patients [33].
In this systematic review, all studies included grade IV gliomas, two studies included
grade II gliomas, and one study included a grade III glioma. The overall survival data
shown in the included studies are not presented according to glioma type. As such, it is
not possible to assess if this variable is affected by the glioma type.
As gliomas have different levels of malignancy, it is expected that the type of tumor
can impact overall survival. Further analysis of this matter is recommended to fully
understand how overall survival varies according to malignancy. Therefore, future large-
scale perspective studies including a greater variety of glioma types are needed.
4.2. Associated Symptomatology
The symptoms associated with KD may involve gastrointestinal problems such as
vomiting, diarrhea, constipation, gastroesophageal reflux, and others such as hypoglycemia,
dizziness, asthenia, hyperketonemia, or metabolic acidosis. Other complications may also
arise, such as dyslipidemia, with hyperlipidemia, hypercholesterolemia, and hypertriglyc-
eridemia; cardiovascular diseases, such as cardiomyopathy; nephrolithiasis, and vitamin
and mineral deficits [3437].
The symptomatology associated with the therapy, or the adverse effects felt by the
patients, were consistent in most of the studies, and the symptom most reported by the pa-
tients was constipation [
8
,
15
17
,
27
,
29
,
30
]. Asthenia was the second most mentioned symp-
tom, being described in studies by Champ et al. [
16
], Klein et al. [
29
],
Panhans et a
l. [
28
],
van der Louw et al., 2019a [10], and Perez et al. [27].
The most expected adverse effect in KD is hypoglycemia, due to the low concentrations
of carbohydrates and the amount of energy provided in the diet [
10
,
15
,
16
,
27
]. In this review,
hypoglycemia was found in four studies, but it promptly resolved with a change in dietary
and medical therapy. Other important symptoms associated with all types of KD were
weight loss, vomiting, diarrhea, and nausea.
Although these symptoms were described during dietary therapy, they were consid-
ered to have a mild and transitory incidence. Some authors considered that there was no
severe symptomatology associated with KD and related the occurrence of serious side
effects to the disease itself and/or medical treatment [15,27,31].
Nutrients 2022,14, 1007 14 of 17
According to the literature, the KD type that has the most side effects is CKD. How-
ever, in this review, it was possible to verify that the symptoms felt by the patients were
transversal, regardless of the diet implemented. Thus, side effects should not be a reason to
select a less restrictive diet.
4.3. Quality of Life
Quality of life is a parameter that must be evaluated with the appropriate objective
and subjective measures to obtain reliable results [
38
]. With the implementation of quality
of life questionnaires, it would be possible to assess this parameter with some certainty.
In this review, we were able to verify that only three studies applied quality of life
questionnaires, namely, Rieger et al., Martin-McGill et al., 2020, and van der Louw et al.,
2019b [
8
,
15
,
30
], and the only validated questionnaires were those used by Martin-McGill
et al., 2020. Although the authors applied these questionnaires, only the study by van der
Louw et al., 2019b presented the results obtained, while Rieger et al. reported qualitative
results and Martin-McGill et al., 2020 mentioned global health status.
The association between the symptoms felt by the patients and their quality of life was
notable. This is supported by Rieger et al., van der Louw et al., 2019b, and Martin-McGill
et al., 2018, who associated a decrease or improvement in quality of life with an increase or
improvement in symptoms, respectively.
In this systematic review, quality of life was a variable related to a high risk of attrition
bias. Therefore, we recommend that other variables should be considered in future inter-
ventions, as these represent a directly dependent factor on the stage of the disease, general
well-being, symptoms, and the patient’s response to therapy. With the results obtained, it
became difficult to establish a relationship between quality of life and the implementation
of KD, so we could only see its inconsistency.
4.4. Study Limitations
This systematic review applied a consistent methodology, with well-defined inclusion
and exclusion criteria, and covering different databases. All studies published to date
were included.
The analysis of articles for the selection of relevant and quality publications was
carried out by two reviewers, reducing the risk of errors in the selection of studies. In cases
of doubt or contradictory opinion, a third element was involved in the selection of studies
to be included.
The present systematic review followed the PRISMA recommendations and was
published in PROSPERO, with the identification CRD42021264173. Cochrane RevMan 5
software, indicated for the development of systematic reviews, was used to analyze the
risk of bias of the included studies. This made it possible to analyze and evaluate these
studies in depth.
Although there is a recent publication that included most of the studies here analyzed,
in the present study, a recent 2021 study (Perez et al.) [
27
] was also analyzed, including
these data in a systematic review for the first time. Additionally, our review is the only one
to date that includes the analysis of side effects, survival, and quality of life.
5. Conclusions
Some limitations were found in the quality of the analyzed data, including the sample
size in the studies (between three and seventeen patients), the different types of gliomas,
and the absence of a control group in most studies. This lack of control makes it difficult to
establish a solid conclusion on the efficacy of KD in the treatment of gliomas.
Since patients must be informed and accept the dietary modifications that KD requires,
performance bias or even the placebo effect could be present. Although the results are
made more robust when interventions are hidden, in the implementation of this kind of
therapy, this is a challenge.
Nutrients 2022,14, 1007 15 of 17
However, in most studies analyzed with the implementation of KD therapy, overall
survival exceeded the prognosis of these patients with the usual chemotherapy therapy
and/or radiotherapy associated with temozolomide. Both the use of different types of KD,
and the presentation of data as a report and not in a more structured way, made it difficult
to compare the side effects felt by patients during the implementation of the different types
of diets.
Furthermore, in the assessment of the quality of life, the use of reported data without
the application of objective and validated measures constituted a limitation to the interpre-
tation of the results. This represented a high risk of friction, and therefore, in most studies,
the data collected were considered as having a high risk of bias. It was not possible to
establish a relationship between quality of life and the implementation of KD. However,
concerning overall survival, in most studies, the prognosis of patients with gliomas who
implemented KD as therapy was higher compared to common therapy.
Although the results obtained point to a positive effect of KD as adjuvant therapy of
malignant gliomas, it is still important to develop new research of high quality that aims to
minimize the risks of bias.
Author Contributions:
Conceptualization, B.S. and A.C.M.; methodology, B.S., A.C.M. and M.L.A.;
software, B.S., M.L.A. and P.A.O.; validation, B.S. and A.C.M.; formal analysis, B.S., A.C.M. and
P.A.O.; investigation, B.S. and A.C.M.; resources, A.C.M. and M.L.A.; data curation, B.S. and P.A.O.;
writing—original draft preparation, B.S.; writing—review and editing, B.S., A.C.M. and P.A.O.;
supervision, A.C.M. All authors have read and agreed to the published version of the manuscript.
Funding: This research received no external funding.
Institutional Review Board Statement: Not applicable.
Informed Consent Statement: Not applicable.
Data Availability Statement:
The protocol for this systematic review was registered in the Inter-
national Prospective Register of Systematic Reviews (PROSPERO) under the registration number
CRD42021264173.
Acknowledgments:
This study was part of the MSc thesis in clinical nutrition of one of the authors
(B.S.), which was supervised by another author (supervisor A.C.M.) from the Faculty of Medicine of
Lisbon and Lisbon School of Health Technology, Lisbon, Portugal. The H&TRC author gratefully
acknowledges the FCT/MCTES national support through UIDB/05608/2020 and UIDP/05608/2020.
Conflicts of Interest: The authors declare no conflict of interest.
References
1.
Artzi, M.; Liberman, G.; Vaisman, N.; Bokstein, F.; Vitinshtein, F.; Aizenstein, O.; Ben Bashat, D. Changes in cerebral metabolism
during ketogenic diet in patients with primary brain tumors: 1H-MRS study. J. Neuro-Oncol.
2017
,132, 267–275. [CrossRef]
[PubMed]
2.
Woodhouse, C.; Ward, T.; Gaskill-Shipley, M.; Chaudhary, R. Feasibility of a modified atkins diet in glioma patients during
radiation and its effect on radiation sensitization. Curr. Oncol. 2019,26, e433–e438. [CrossRef] [PubMed]
3.
Poff, A.; Koutnik, A.P.; Egan, K.M.; Sahebjam, S.; D’Agostino, D.; Kumar, N.B. Targeting the Warburg effect for cancer treatment:
Ketogenic diets for management of glioma. Semin. Cancer Biol. 2019,56, 135–148. [CrossRef]
4.
Schwartz, K.A.; Noel, M.; Nikolai, M.; Chang, H.T. Investigating the ketogenic diet as treatment for primary aggressive brain
cancer: Challenges and lessons learned. Front. Nutr. 2018,5, 11. [CrossRef] [PubMed]
5.
Foppiani, A.; De Amicis, R.; Lessa, C.; Leone, A.; Ravella, S.; Ciusani, E.; Silvani, A.; Zuccoli, G.; Battezazati, A.; Lamperti, E.; et al.
Isocaloric Ketogenic Diet in Adults with High-Grade Gliomas: A Prospective Metabolic Study. Nutr. Cancer
2021
,73, 1004–1014.
[CrossRef] [PubMed]
6. Vasconelos, C.; Rola, M. Dieta Cetogénica—Abordagem Nutricional. Rev. Nutrícias 2014,22, 16–19.
7.
Van der Louw, E.; van den Hurk, D.; Neal, E.; Leiendecker, B.; Fitzsimmon, G.; Dority, L.; Thompson, L.; Marchio, M.; Dudzinska,
M.; Dressler, A.; et al. Ketogenic diet guidelines for infants with refractory epilepsy. Eur. J. Paediatr. Neurol.
2016
,20, 798–809.
[CrossRef]
8.
Rieger, J.; Bähr, O.; Maurer, G.D.; Hattingen, E.; Franz, K.; Brucker, D.; Walenta, S.; Kammerer, U.; Coy, J.F.; Waller, M.; et al.
ERGO: A pilot study of ketogenic diet in recurrent glioblastoma. Int. J. Oncol. 2014,45, 1843–1852. [CrossRef]
Nutrients 2022,14, 1007 16 of 17
9.
Vidali, S.; Aminzadeh, S.; Lambert, B.; Rutherford, T.; Sperl, W.; Kofler, B.; Feichtinger, R.G. Mitochondria: The ketogenic diet—A
metabolism-based therapy. Int. J. Biochem. Cell Biol. 2015,63, 55–59. [CrossRef]
10.
Van der Louw, E.J.T.M.; Reddingius, R.E.; Olieman, J.F.; Neuteboom, R.F.; Catsman-Berrevoets, C.E. Ketogenic diet treatment
in recurrent diffuse intrinsic pontine glioma in children: A safety and feasibility study. Pediatric Blood Cancer
2019
,66, e27561.
[CrossRef]
11.
Woolf, E.C.; Scheck, A.C. The ketogenic diet for the treatment of malignant glioma. J. Lipid Res.
2015
,56, 5–10. [CrossRef]
[PubMed]
12. Morris, A.A.M. Cerebral ketone body metabolism. J. Inherit. Metab. Dis. 2005,28, 109–121. [CrossRef] [PubMed]
13.
Ebrahimpour-Koujan, S.; Shayanfar, M.; Benisi-Kohansal, S.; Mohammad-Shirazi, M.; Sharifi, G.; Esmaillzadeh, A. Adherence to
low carbohydrate diet in relation to glioma: A case-control study. Clin. Nutr. 2019,38, 2690–2695. [CrossRef] [PubMed]
14. Perry, A.; Wesseling, P. Histologic classification of gliomas. Handb. Clin. Neurol. 2016,134, 71–95. [PubMed]
15.
Van der Louw, E.J.T.M.; Olieman, J.F.; van den Bemt, P.M.; Bromberg, J.E.; Oomen-de Hoop, E.; Neuteboom, R.F.; Catsman-
Berrevoets, C.E.; Vincent, A.J. Ketogenic diet treatment as adjuvant to standard treatment of glioblastoma multiforme: A feasibility
and safety study. Ther. Adv. Med. Oncol. 2019,11, 1758835919853958N. [CrossRef]
16.
Champ, C.E.; Palmer, J.D.; Volek, J.S.; Werner-Wasik, M.; Andrews, D.W.; Evans, J.J.; Glass, J.; Kim, L.; Shi, W. Targeting
metabolism with a ketogenic diet during the treatment of glioblastoma multiforme. J. Neuro-Oncol.
2014
,117, 125–131. [CrossRef]
17.
Martin-McGill, K.J.; Marson, A.G.; Smith, C.T.; Jenkinson, M.D. The modified ketogenic diet in adults with glioblastoma: An
evaluation of feasibility and deliverability within the national health service. Nutr. Cancer 2018,70, 643–649. [CrossRef]
18.
Woolf, E.C.; Syed, N.; Scheck, A.C. Tumor metabolism, the ketogenic diet and
β
-hydroxybutyrate: Novel approaches to adjuvant
brain tumor therapy. Front. Mol. Neurosci. 2016,9, 122. [CrossRef]
19.
Winter, S.F.; Loebel, F.; Dietrich, J. Role of ketogenic metabolic therapy in malignant glioma: A systematic review. Crit. Rev. Oncol.
Hematol. 2017,112, 41–58. [CrossRef]
20. Wen, P.Y.; Kesari, S. Malignant Gliomas in Adults. 2008. Available online: www.nejm.org (accessed on 8 May 2021).
21.
Louis, D.N.; Ohgaki, H.; Wiestler, O.D.; Cavenee, W.K.; Burger, P.C.; Jouvet, A.; Scheithauer, W.; Kleihues, P. The 2007 WHO
classification of tumours of the central nervous system. Acta Neuropathol. 2007,114, 97–109. [CrossRef]
22.
Strowd, R.E.; Grossman, S.A. The role of glucose modulation and dietary supplementation in patients with central nervous
system tumors. Curr. Treat. Options Oncol. 2015,16, 36. [CrossRef] [PubMed]
23.
Moher, D.; Tetzlaff, J.; Altman, D.G. Preferred reporting items for systematic reviews and meta-analyses: The PRISMA statement.
PLoS Med. 2009,6, 332–336. [CrossRef] [PubMed]
24.
Guidance Notes for Registering a Systematic Review Protocol with PROSPERO. 2016. Available online: www.crd.york.ac.uk/
prospero (accessed on 8 May 2021).
25.
Ouzzani, M.; Hammady, H.; Fedorowicz, Z.; Elmagarmid, A. Rayyan—A web and mobile app for systematic reviews. Syst. Rev.
2016,5, 210. [CrossRef] [PubMed]
26. Review Manager (RevMan), Version 5.4.1; The Cochrane Collaboration: London, UK, 2020.
27.
Perez, A.; van der Louw, E.; Nathan, J.; El-Ayadi, M.; Golay, H.; Korff, C.; Ansari, M.; Catsman-Berrevoets, C.; von Bueren, A.O.
Ketogenic diet treatment in diffuse intrinsic pontine glioma in children: Retrospective analysis of feasibility, safety, and survival
data. Cancer Rep. 2021,4, e1383. [CrossRef] [PubMed]
28.
Panhans, C.M.; Gresham, G.; Amaral, J.L.; Hu, J. Exploring the feasibility and effects of a ketogenic diet in patients with CNS
malignancies: A retrospective case series. Front. Neurosci. 2020,14, 390.
29.
Klein, P.; Tyrlikova, I.; Zuccoli, G.; Tyrlik, A.; Maroon, J.C. Treatment of glioblastoma multiforme with “classic” 4:1 ketogenic diet
total meal replacement. Cancer Metabol. 2020,8, 682243. [CrossRef]
30.
Martin-McGill, K.J.; Marson, A.G.; Tudur Smith, C.; Young, B.; Mills, S.J.; Cherry, M.G.; Jenkinson, M.D. Ketogenic diets as
an adjuvant therapy for glioblastoma (KEATING): A randomized, mixed methods, feasibility study. J. Neuro-Oncol.
2020
,147,
213–227. [CrossRef]
31.
Schwartz, K.; Chang, H.T.; Nikolai, M.; Pernicone, J.; Rhee, S.; Olson, K.; Kurniali, P.C.; Hord, N.G.; Noel, M. Treatment of glioma
patients with ketogenic diets: Report of two cases treated with an IRB-approved energy-restricted ketogenic diet protocol and
review of the literature. Cancer Metab. 2015,3, 3. [CrossRef]
32.
Zuccoli, G.; Marcello, N.; Pisanello, A.; Servadei, F.; Vaccaro, S.; Mukherjee, P.; Seyfried, T.N. Metabolic management of
glioblastoma multiforme using standard therapy together with a restricted ketogenic diet: Case report. Nutr. Metab.
2010
,
7, 33
.
[CrossRef]
33.
Noorlag, L.; de Vos, F.Y.; Kok, A.; Broekman, M.L.; Seute, T.; Robe, P.A.; Snijders, T.J. Treatment of malignant gliomas with
ketogenic or caloric restricted diets: A systematic review of preclinical and early clinical studies. Clin. Nutr.
2019
,38, 1986–1994.
[CrossRef]
34.
Mosek, A.; Natour, H.; Neufeld, M.Y.; Shiff, Y.; Vaisman, N. Ketogenic diet treatment in adults with refractory epilepsy: A
prospective pilot study. Seizure 2009,18, 30–33. [CrossRef] [PubMed]
35. Kossoff, E.; Wang, H.S. Dietary therapies for epilepsy. Biomed. J. 2013,36, 2–8. [CrossRef] [PubMed]
Nutrients 2022,14, 1007 17 of 17
36.
Bergqvist, A.G.C. Long-term monitoring of the ketogenic diet: Do’s and don’ts. Epilepsy Res.
2012
,100, 261–266. [CrossRef]
[PubMed]
37. Sampaio, L.P.d. Ketogenic diet for epilepsy treatment. Arq. Neuro-Psiquiatr. 2016,74, 842–848. [CrossRef]
38. Martin-McGill, K.J.; Srikandarajah, N.; Marson, A.G.; Smith, C.T.; Jenkinson, M.D. The role of ketogenic diets in the therapeutic
management of adult and paediatric gliomas: A systematic review. CNS Oncol. 2018,7, CNS17. [CrossRef]
... These processes result in reduced tumour growth, improved cancer survival rates, and increased effectiveness of chemotherapy and radiotherapy [29]. The KD has shown promising results as an additional treatment for gliomas [30,31]. In this scenario, drug interactions may arise in patients diagnosed with cerebral tumours who are also receiving pharmacologic treatment for other conditions. ...
... These proces result in reduced tumour growth, improved cancer survival rates, and increased effect ness of chemotherapy and radiotherapy [29]. The KD has shown promising results as additional treatment for gliomas [30,31]. In this scenario, drug interactions may aris patients diagnosed with cerebral tumours who are also receiving pharmacologic tr ment for other conditions. ...
... The effects of the KD on serum electrolytes and acid-base status, the impact of intravenous fluid selection on acid-base status, the modification of ketogenic status through the administration of glucose in intravenous fluids or medication, the risk of hypoglycaemia, and the effects of ketosis and acidosis on cardiovascular function are among the specific perioperative concerns of patients undergoing the KD [30]. ...
Article
Full-text available
It is widely acknowledged that the ketogenic diet (KD) has positive physiological effects as well as therapeutic benefits, particularly in the treatment of chronic diseases. Maintaining nutritional ketosis is of utmost importance in the KD, as it provides numerous health advantages such as an enhanced lipid profile, heightened insulin sensitivity, decreased blood glucose levels, and the modulation of diverse neurotransmitters. Nevertheless, the integration of the KD with pharmacotherapeutic regimens necessitates careful consideration. Due to changes in their absorption, distribution, metabolism, or elimination, the KD can impact the pharmacokinetics of various medications, including anti-diabetic, anti-epileptic, and cardiovascular drugs. Furthermore, the KD, which is characterised by the intake of meals rich in fats, has the potential to impact the pharmacokinetics of specific medications with high lipophilicity, hence enhancing their absorption and bioavailability. However, the pharmacodynamic aspects of the KD, in conjunction with various pharmaceutical interventions, can provide either advantageous or detrimental synergistic outcomes. Therefore, it is important to consider the pharmacokinetic and pharmacodynamic interactions that may arise between the KD and various drugs. This assessment is essential not only for ensuring patients’ compliance with treatment but also for optimising the overall therapeutic outcome, particularly by mitigating adverse reactions. This highlights the significance and necessity of tailoring pharmacological and dietetic therapies in order to enhance the effectiveness and safety of this comprehensive approach to managing chronic diseases.
... On the other hand, a restricted ketogenic diet (RKD) may offer an effective and safe treatment adjunct in patients with GBM [12][13][14][15][16][17][18][19][20][21]. Its use is based on the concept that brain cancer is partly a metabolic disorder, which could be controlled by depriving cancer cells of essential metabolic substrates, such as glucose and glutamine [12][13][14][15][16][17][18][19][20][21]. ...
... On the other hand, a restricted ketogenic diet (RKD) may offer an effective and safe treatment adjunct in patients with GBM [12][13][14][15][16][17][18][19][20][21]. Its use is based on the concept that brain cancer is partly a metabolic disorder, which could be controlled by depriving cancer cells of essential metabolic substrates, such as glucose and glutamine [12][13][14][15][16][17][18][19][20][21]. This metabolic effect is achieved by decreasing the intake of calories and carbohydrates while increasing the relative proportion of lipids in the diet [12][13][14][15][16][17][18][19][20][21]. ...
... Its use is based on the concept that brain cancer is partly a metabolic disorder, which could be controlled by depriving cancer cells of essential metabolic substrates, such as glucose and glutamine [12][13][14][15][16][17][18][19][20][21]. This metabolic effect is achieved by decreasing the intake of calories and carbohydrates while increasing the relative proportion of lipids in the diet [12][13][14][15][16][17][18][19][20][21]. Evidence from in vitro studies shows that a RKD has an anti-angiogenic, anti-invasive, anti-inflammatory effect on GBM cells [12][13][14][15][16][17][18][19][20][21]. ...
Article
Full-text available
Glioblastoma is a highly aggressive brain tumor that has a poor prognosis despite various treatments like surgery, chemotherapy, and irradiation. However, a restricted ketogenic diet (RKD), which has been proven to be effective in treating drug-resistant epilepsy, could be a potential adjunct in the treatment of certain GBM cases. Our study aimed to highlight the existing knowledge, identify collaboration networks, and emphasize the ongoing research based on highly cited studies. During the literature search, we found 119 relevant articles written between 2010 and 2023. Among the top 20 most cited articles, there were seven laboratory and five clinical studies. The works of Olson LK, Chang HT, Schwartz KA, and Nikolai M from the Michigan State University, followed by Seyfried TN and Mukherjee P from Boston College, and Olieman JF, and Catsman-Berrevoets CE from the University Medical Center of Rotterdam, were significant contributions. The laboratory studies showed that RKD had a significant antitumor effect and could prolong survival in mouse glioblastoma models. The clinical studies verified the tolerability, efficacy, and safety of RKD in patients with GBM, but raised concerns about whether it could be used as a single therapy. The current research interest is focused on the efficacy of using RKD as an adjunct in selected chemotherapy regimens and demonstrates that it could provide GBM patients with better treatment options.
... The most reported symptoms are constipation and asthenia. Hypoglycemia is the most anticipated adverse effect; however, mild hypoglycemia can be intended for therapeutic interventions (34). Because cancer cells lack metabolic flexibility due to their mitochondrial mutations and abnormalities, this hypoglycemic state can aggravate oxidative cellular stress. ...
... Ketone bodies can also promote ROS production in cancer cells. However, there are common side effects of ketogenic diets, including constipation, asthenia, and hypoglycemia (34). HBOT and HT are other stressors for cancer cells that are known to work synergistically with a ketogenic diet. ...
... For example, ketosis can slow glioblastoma tumor formation because gliomas are unable to effectively use ketones, and this induces a direct Warburg effect. 31 However, epithelial cancer cells of the head and neck metabolize ketones and lactate efficiently, and some research shows that a ketogenic state might propagate the growth of these cancers. 32 Ketone bodies may be usable by the actively growing and stem cellelike component of tumors such as breast cancer 33 ; therefore, we need to better understand the long-term effects of a ketotic state in cancer. ...
Article
Full-text available
Lifestyle factors and their impact on cancer prevention, prognosis, and survivorship are increasingly recognized in the medical literature. Lifestyle factors are primarily defined here as diet and physical activity. We conducted a narrative review of the primary published data, including randomized controlled trials and prospective studies, on the impact of primary lifestyle factors on oncogenesis and clinical outcomes in the preventative and survivorship setting. First, we discuss the oncogenic mechanisms behind primary lifestyle factors (diet, physical activity and, within these 2, obesity). Then, we discuss the impact of adherence to lifestyle guidelines and dietary patterns on cancer incidence based on primary data. Owing to the plethora of published literature, to summarize the data in a more efficient manner, we describe the role of physical activity on cancer incidence using summative systematic reviews. We end by synthesizing the primary data on lifestyle factors in the survivorship setting and conclude with potential future directions. In brief, the various large-scale studies investigating the role diet and physical activity have reported a beneficial effect on cancer prevention and survivorship. Although the impact of single lifestyle factors on cancer incidence risk reduction is generally supported, holistic approaches to address the potential synergistic impact of multiple lifestyle factors together in concert is limited. Future research to identify the potentially synergistic effects of lifestyle modifications on oncogenesis and clinical outcomes is needed, particularly in cancer subtypes beyond colorectal and breast cancers.
Article
Cancer is a global health concern influenced by genetics, environment and lifestyle choices. Recent research shows that a ketogenic diet (KD) might ease cancer symptoms and reduce tumour size. We hypothesised that the KD could result in improvements in cancer‐related variables. Therefore, this study aims to perform a systematic review and meta‐analysis to assess the KD's efficacy for patients with cancer. The databases PubMed (MEDLINE), Web of Science, CINAHL and Open Grey were utilised for conducting a systematic review and meta‐analysis. The analysis was limited to randomised controlled trials with adult participants aged 18 years and above. Levels of glucose, cholesterol, insulin‐like growth factor 1, weight and quality of life were evaluated following the KD. After identifying 596 articles in the initial search, eight studies, lasting between 4 and 16 weeks, were included in the systematic review and seven in the meta‐analysis. The KD led to decreased glucose levels in patients with cancer but did not show significant improvements in cholesterol, insulin‐like growth factor 1, weight or quality of life. Based on the results of this systematic review and meta‐analysis, there is insufficient evidence to establish a definitive link between the KD and cancer‐related parameters. While some studies suggest potential benefits in terms of some outcomes and tumour size reduction, further research is required to fully comprehend the effects of this diet.
Article
As treatment for glioma advances, with an attendant improvement in length of patient survival, the quality of that survival has rightly become an increasingly important patient-centered metric, and health-related quality of life (HRQOL) an important outcome measure. HRQOL is a self-assessed, multidimensional concept encompassing the physical, emotional, and social components of quality of life associated with illness and its treatment. Neurosurgeons caring for patients with gliomas should be aware of the latest research on HRQOL to understand mechanisms by which it can be improved. Neurosurgical outcomes related to surgical complications and neurological deficits can be important determinants of HRQOL and are well understood by neurosurgeons. However, an understanding of more general or global determinants of HRQOL not commonly addressed in the clinic, and implementation of the attendant evidence-based interventions to address them, would be transformative. The authors explore HRQOL determinants related to patient-, social-, tumor-, and treatment-related factors, with a particular emphasis on the strongest determinants, fatigue, sleep disturbance, anxiety, depression, neurocognitive dysfunction, caregiver distress, and end-of-life concerns. Evidence-based interventions are reviewed, including fatigue management, cognitive rehabilitation, insomnia interventions exercise, caregiver training, palliative care, and an overall multidisciplinary team approach. Lastly, features of a program are outlined that would embed HRQOL in neurosurgical care to the benefit of both patients and staff.
Article
Full-text available
Background Prior systematic reviews addressing the impact of diet on cancer outcomes have focused on specific dietary interventions. In this systematic review, we assessed all RCTs investigating dietary interventions for cancer patients, examining the range of interventions, endpoints, patient populations, and results. Methods This systematic review identified all RCTs conducted prior to January 2023 testing dietary interventions in patients with cancer. Assessed outcomes included quality of life, functional outcomes, clinical cancer measurements (eg, progression-free survival, response rates), overall survival, and translational endpoints (eg, inflammatory markers). Results 252 RCTs were identified involving 31,067 patients. The median sample size was 71 (interquartile range 41 to 118), and 80 (32%) studies had a sample size greater than 100. Most trials (n = 184, 73%) were conducted in the adjuvant setting. Weight/body composition and translational endpoints were the most common primary endpoints (n = 64, 25%; n = 52, 21%, respectively). Direct cancer measurements and overall survival were a primary endpoint in 20 (8%) and seven (3%) studies respectively. Eight trials with a primary endpoint of cancer measurement (40%) met their endpoint. Large trials in colon (n = 1429), breast (n = 3088), and prostate cancer (n = 478) each showed no effect of dietary interventions on endpoints measuring cancer. Conclusion Most RCTs of dietary interventions in cancer are small and measure non-clinical endpoints. Although only a small number of large RCTs have been conducted to date, these trials have not shown an improvement in cancer outcomes. Currently, there is limited evidence to support dietary interventions as a therapeutic tool in cancer care.
Article
Introdução: Um dos fatores dietéticos relacionados à recuperação de pacientes oncológicos que vem se destacando é a dieta cetogênica (DC). Devido ao metabolismo glicolítico predominante das células cancerígenas pressupõe que a restrição de carboidratos pode suprimir ou retardar a atividade do câncer. Objetivo: Realizar um levantamento sobre a influência da DC no tratamento de pacientes oncológicos. Materiais e métodos: Trata-se de uma revisão de literatura do tipo integrativa, na qual foram consultados artigos científicos das bases de dados: BVS, PUBMED e SCIELO, publicados nos últimos 5 anos (2017 a 2022). A pesquisa ocorreu por meio de palavras consultadas nos Descritores em Ciências da Saúde (DeCs), utilizando as combinações dos termos: Dieta Cetogênica/Ketogenic Diet AND Câncer/cancer OR Neoplasias/Neoplasms. Discussão: Encontrou-se um total de 320 artigos de acordo com os descritores relacionados ao tema. Após a leitura de títulos, resumos e inclusão dos filtros, foi realizado o processo de refinamento dos artigos, permanecendo um total de 7 artigos na composição desse estudo. Foram encontradas evidências de que as dietas cetogênicas (DCs) são capazes de induzir mudanças positivas profundas no peso e composição corporal, juntamente com os efeitos das terapias locais e sistêmicas, importantes preditores de evolução do câncer. Conclusões: Os resultados sugerem que as DCs sejam seguras na população oncológica. No entanto, evidências clínicas mais amplas e consistentes são necessárias para minimizar os riscos de viés e para determinar a eficácia da DC como uma terapia adjuvante não farmacológica no tratamento do câncer.
Article
Full-text available
Background Diffuse intrinsic pontine glioma (DIPG) is one of the most devastating diseases among children with cancer, thus novel strategies are urgently needed. Aims We retrospectively evaluated DIPG patients exposed to the carbohydrate restricted ketogenic diet (KD) with regard of feasibility, safety, and overall survival (OS). Methods and results Searches of MEDLINE and Embase identified five hits meeting the search criteria (diagnosis of DIPG and exposure to KD). One additional case was identified by contact with experts. Individual patient data were extracted from publications or obtained from investigators. The inclusion criteria for analysis of the data were defined as DIPG patients who were exposed to the KD for ≥3 months. Feasibility, as described in the literature, was the number of patients able to follow the KD for 3 months out of all DIPG patients identified. OS was estimated by the Kaplan‐Meier method. Five DIPG patients (males, n = 3; median age 4.4 years; range, 2.5‐15 years) meeting the inclusion criteria were identified. Analysis of the available data suggested that the KD is generally relatively well tolerated. Only mild gastro‐intestinal complaints, one borderline hypoglycemia (2.4 mmol/L) and one hyperketosis (max 7.2 mmol/L) were observed. Five out of six DIPG patients identified adhered for ≥3 months (median KD duration, 6.5 months; range, 0.25‐2 years) to the diet. The median OS was 18.7 months. Conclusion Our study provides evidence that it may be feasible for pediatric DIPG patients to adhere for at least 3 months to KD. In particular cases, diet modifications were done. The clinical outcome and OS appear not to be impacted in a negative way. KD might be proposed as adjuvant therapy when large prospective studies have shown feasibility and safety. Future studies might ideally assess the impact of KD on clinical outcome, quality of life, and efficacy.
Article
Full-text available
Introduction Glioblastoma (GBM) has poor survival with standard treatment. Experimental data suggest potential for metabolic treatment with low carbohydrate ketogenic diet (KD). Few human studies of KD in GBM have been done, limited by difficulty and variability of the diet, compliance, and feasibility issues. We have developed a novel KD approach of total meal replacement (TMR) program using standardized recipes with ready-made meals. This pilot study evaluated feasibility, safety, tolerability, and efficacy of GBM treatment using TMR program with “classic” 4:1 KD. Method GBM patients were treated in an open-label study for 6 months with 4:1 [fat]:[protein + carbohydrate] ratio by weight, 10 g CH/day, 1600 kcal/day TMR. Patients were either newly diagnosed (group 1) and treated adjunctively to radiation and temozolomide or had recurrent GBM (group 2). Patients checked blood glucose and blood and urine ketone levels twice daily and had regular MRIs. Primary outcome measures included retention, treatment-emergent adverse events (TEAEs), and TEAE-related discontinuation. Secondary outcome measures were survival time from treatment initiation and time to MRI progression. Results Recruitment was slow, resulting in early termination of the study. Eight patients participated, 4 in group 1 and 4 in group 2. Five (62.5%) subjects completed the 6 months of treatment, 4/4 subjects in group 1 and 1/4 in group 2. Three subjects stopped KD early: 2 (25%) because of GBM progression and one (12.5%) because of diet restrictiveness. Four subjects, all group 1, continued KD on their own, three until shortly before death, for total of 26, 19.3, and 7 months, one ongoing. The diet was well tolerated. TEAEs, all mild and transient, included weight loss and hunger (n = 6) which resolved with caloric increase, nausea (n = 2), dizziness (n = 2), fatigue, and constipation (n = 1 each). No one discontinued KD because of TEAEs. Seven patients died. For these, mean (range) survival time from diet initiation was 20 months for group 1 (9.5–27) and 12.8 months for group 2 (6.3–19.9). Mean survival time from diagnosis was 21.8 months for group 1 (11–29.2) and 25.4 months for group 2 ( 13.9–38.7). One patient with recurrent GBM and progression on bevacizumab experienced a remarkable symptom reversal, tumor shrinkage, and edema resolution 6–8 weeks after KD initiation and survival for 20 months after starting KD. Conclusion Treatment of GBM patients with 4:1 KD using total meal replacement program with standardized recipes was well tolerated. The small sample size limits efficacy conclusions. Trial registration NCT01865162 registered 30 May 2013, and NCT02302235 registered 26 November 2014, https://clinicaltrials.gov/
Article
Full-text available
The use of the ketogenic diet (KD) as an adjuvant therapy in high-grade gliomas (HGG) is supported by preclinical studies, but clinical data on its effects on metabolism are currently lacking. In this study, we describe the effects of a KD on glucose profile, ketonemia, energy metabolism, and nutritional status, in adults affected by HGG. This was a single-arm prospective study. An isocaloric 3:1 KD was administered for 1 mo. Glucose profile was assessed by using fasting glycemia, insulin, and glycated hemoglobin. To evaluate ketonemia changes, a hand-held ketone meter was used from home. Energy metabolism was assessed by indirect calorimetry. Nutritional status was evaluated through changes in body composition and in lipid and hepatic profile. No changes in fasting glycemia were observed; however, insulinemia dropped to half of baseline levels. The KD shifted the metabolism, rising ketonemia and decreasing glucose oxidation rate to a quarter of the initial values. Moreover, the KD was generally safe. One-month intervention with the KD was able to act upon key metabolic substrates potentially involved in HGG metabolism. The lack of a significant reduction in fasting glycemia should be investigated in future studies.
Article
Full-text available
Background: Recently, the ketogenic diet has been proposed as an adjunct treatment for a range of medical conditions including weight loss, diabetes, cancer, and neurodegenerative diseases. Because malignant CNS tumors are highly dependent on glucose, the use of a ketogenic diet as an adjunct therapy is currently being explored. This case series summarizes our experience implementing a ketogenic diet for patients with CNS malignancies. Methods: Patients diagnosed with CNS malignancies following a ketogenic diet were identified between 2015 and 2017. Malignancies included confirmed diagnoses of glioblastoma (GBM), astrocytoma, or oligodendroglioma. With guidance from a registered dietitian, ketone levels, glucose levels, and weight were regularly collected for several patients along with patient-reported symptoms and adverse effects. Interested patients were asked to follow a 3:1 ketogenic diet for 120 days. The ketogenic diet is a high-fat, moderate protein, and very low carbohydrate diet, where patients limited carbohydrate intake to ≤20 g per day. Brain imaging was reviewed. A series of descriptive analyses were conducted. Results: The ketogenic diet was initiated in 12 patients of which 8 patients contributed data on their blood glucose and ketone levels. The majority of patients were male (n = 10) with a median age of 45 (range 32–62). Diagnoses included GBM (n = 6), grade 2/3 astrocytomas (n = 5) and one patient with a grade 2 spinal cord astrocytoma. Ten of the 12 patients were receiving concurrent treatment; two received supportive care only. The majority of patients with evaluable data (n = 8) maintained ketone levels above 0.5 mM for the duration of 120-day period. Ketone levels generally increased from baseline while glucose levels and BMI decreased. Overall, patients reported improved symptoms over the course of the diet. Imaging also suggested improved disease control and reduction in vasogenic edema. Conclusion: Taking advantage of a tumor’s metabolic inflexibility can have a positive impact on patients, particularly those with CNS malignancies. More structured and statistically planned clinical trials are needed to determine the margin of impact of a ketogenic diet.
Article
Full-text available
Purpose We conducted a feasibility study to investigate the use of ketogenic diets (KDs) as an adjuvant therapy for patients with glioblastoma (GBM), investigating (i) trial feasibility; (ii) potential impacts of the trial on patients’ quality of life and health; (iii) patients’ perspectives of their decision-making when invited to participate in the trial and (iv) recommending improvements to optimize future phase III trials. Methods A single-center, prospective, randomized, pilot study (KEATING), with an embedded qualitative design. Twelve newly diagnosed patients with GBM were randomized 1:1 to modified ketogenic diet (MKD) or medium chain triglyceride ketogenic diet (MCTKD). Primary outcome was retention at three months. Semi-structured interviews were conducted with a purposive sample of patients and caregivers (n = 15). Descriptive statistics were used for quantitative outcomes and qualitative data were analyzed thematically aided by NVivo. Results KEATING achieved recruitment targets, but the recruitment rate was low (28.6%). Retention was poor; only four of 12 patients completed the three-month diet (MCTKD n = 3; MKD n = 1). Participants’ decisions were intuitive and emotional; caregivers supported diet implementation and influenced the patients’ decision to participate. Those who declined made a deliberative and considered decision factoring diet burden and quality of life. A three-month diet was undesirable to patients who declined and withdrew. Conclusion Recruitment to a KD trial for patients with GBM is possible. A six-week intervention period is proposed for a phase III trial. The role of caregivers should not be underestimated. Future trials should optimize and adequately support the decision-making of patients.
Article
Full-text available
Background: Gliomas are the most dreaded primary brain tumour because of their dismal cure rates. Ketogenic-type diets (kds) are high-fat, low-protein, and low-carbohydrate diets; the modified Atkins diet (mad) is a less-stringent version of a kd that still generates serum ketones in patients. The purpose of the present study was to retrospectively examine the feasibility of attaining ketosis and the safety of the mad in patients undergoing radiation and chemotherapy treatment for glioma. The rate of pseudoprogression (psp) after treatment was also assessed as a marker of radiation sensitization. To our knowledge, this dataset is the largest published relating to patients with glioma undergoing kd during radiation and chemotherapy. Methods: We retrospectively studied 29 patients with grades ii-iv astrocytoma following the mad during standard radiation and chemotherapy. Feasibility of attaining ketosis was assessed though levels of beta hydroxybutyrate in blood. Pre- and post-radiation magnetic resonance images were evaluated for psp by a neuroradiologist blinded to patient data. Results: In the 29 patients who started the mad during radiation, ketosis was achieved in all 29 (100%). No serious adverse events occurred secondary to the mad. Of those 29 patients, 19 had glioblastoma multiforme. Of the latter 19 patients, 11 (58%) showed psp after mad and radiation and temozolomide therapy. Conclusions: A modified Atkins diet is feasible and safe for glioma patients during radiation and chemotherapy treatment. The mad and resulting ketosis could play a role as a radiation sensitizer.
Article
Full-text available
Background: High-grade glioma cells consume mainly glucose and cannot compensate for glucose restriction. Apoptosis may potentially occur under carbohydrate restriction by a ketogenic diet (KD). We explored the feasibility and safety of KD during standard treatment of chemoradiation in patients with glioblastoma multiforme. Methods: A full liquid KD induced ketosis within 2 weeks before start of chemoradiation. After 6 weeks, the KD was modified with solid foods and medium-chain-triglyceride emulsions and used for an additional 6 weeks while maintaining ketosis. During the total study period (14 weeks), feasibility, safety, coping (both patient and partner), quality of life (QoL), neurological functioning and impairment were measured. Overall survival was analyzed with actuarial estimates. Results: Eleven patients started the study protocol, nine reached ketosis and six (67%) completed the study. Severe adverse effects did not occur. The majority of coping scores ranged from 3 to 6 on a 10-point scale at all timepoints; QoL, neurological functioning, and impairment did not essentially change over time; overall survival ranged between 9.8 and 19.0 months. Conclusion: KD was feasible and safe as an adjuvant to standard chemoradiation treatment of glioblastoma multiforme. A supportive partner and intensive counseling were essential for coping. Future research should identify possible beneficial effects on overall survival. Clinical trial registration: Netherlands Trial Registry: NTR5167 (registration date 29-01-2015), http://www.trialregister.nl/trialreg/index.asp.
Article
Full-text available
1 Background The mean overall survival rate of children with diffuse intrinsic pontine glioma (DIPG) is 9–11 months, with current standard treatment with fractionated radiotherapy and adjuvant chemotherapy. So far, novel therapeutic strategies have not yet resulted in significantly better survival. The main source of energy for glioblastoma cells is glucose. Therefore, metabolic alterations induced by the use of the extremely carbohydrate‐restricted ketogenic diet (KD) as adjuvant therapy are subject of interest in cancer research. 2 Procedure This study explores the safety and feasibility of the KD in children with recurrent DIPG and no remaining treatment options. Safety was defined as the number of adverse effects. Feasibility was defined as the number of patients who were able to use the KD for three months. Coping of patients and parents was measured with questionnaires. 3 Results Three of 14 children referred to our hospital between 2010 and 2015 were included. Two patients completed the study, and one died before the end of the study. Hospitalizations were needed for placing a nasogastric tube (n = 1) and epileptic seizures (n = 1). Adverse effects related to the diet were mild and transient. Parents were highly motivated during the study. 4 Conclusion Use of KD is safe and feasible, but the effect on survival has to be proven in a larger cohort of children who start the KD earlier after diagnosis, preferably as adjuvant therapy to fractionated radiotherapy.
Article
Background: Observational studies that investigated the association between adherence to low carbohydrate diet and odds of glioma are sparse. This is particularly relevant in developing countries where dietary carbohydrate intake is very high. Objective: This study was, therefore, conducted to investigate the association between adherence to low carbohydrate dietary pattern and odds of glioma among Iranian adults. Methods: In this hospital-based case-control study, 128 newly diagnosed glioma cases were recruited and 256 age- and sex-matched subjects were enrolled as controls. Dietary intakes were examined by the use of a 126-item validated FFQ. Low carbohydrate diet (LCD) score was computed for each participant based on deciles of percentages of energy from carbohydrates, proteins, and fats. Individuals in the lowest decile of carbohydrate consumption received 10 points. Participants in second decile received 9 points and so on down to participants in the highest decile received 1 points. For fat and protein intakes, the points assigned to deciles were reversed; such that those in the highest decile received 10 points and those in the lowest decile received 1 point. We then summed all points of the three macronutrients to achieve the overall diet score, which ranged from 3 (highest carbohydrate intake and lowest fat and protein intakes) to 30 (lowest carbohydrate intake and highest fat and protein intakes). The higher the score, the greater adherence to the LCD dietary pattern. Results: After adjustment for age and sex, we found that individuals in top tertile of LCD score were 60% less likely to have glioma compared with those in the bottom tertile (OR: 0.40; 95% CI: 0.23, 0.67). Additional controlling for other potential confounders made the association slightly attenuated (OR: 0.49; 95%CI: 0.26, 0.93). Further adjustment for dietary nutrient intakes strengthened the association (OR: 0.32; 95%CI: 0.12, 0.81). After additional controlling for BMI, we found that adherence to LCD was protectively associated with reduced odds of glioma (OR: 0.32; 95%CI: 0.12, 0.81). Discussion: We found an inverse association between adherence to LCD and odds of glioma among Iranian population. Prospective cohort studies are needed to confirm these findings.
Article
Background & aims: Patients with malignant gliomas have a poor prognosis. Diets that lower blood glucose, such as ketogenic or caloric restricted diets (KCRDs), are hypothesized to reduce tumor growth and improve survival. In this systematic review, we summarize preclinical and clinical data on KCRDs in gliomas. Methods: We searched PubMed and Embase for preclinical and clinical studies on KCRDs in gliomas, and extracted data on surrogate and clinically relevant endpoints, in accordance with PRISMA statement. Quality assessment of clinical studies was performed with use of Cochrane Collaboration's tool. We performed Fisher's exact test to examine associations between surrogate and clinically relevant endpoints. Results: We included 24 preclinical studies, seven clinical studies and one mixed study. Both preclinical and clinical studies were highly heterogeneous. Preclinically, KCRDs reduced tumor growth, but only a small majority of the in vivo studies found improved survival. These effects were stronger in groups with decreased blood glucose than in those with increased ketones, and also when other therapies were used concomitantly. Finally, KCRDs influence multiple molecular-biological pathways, including the PTEN/Akt/TSC2 and NF-kB pathway. In clinical studies, KCRDs seem to be safe and feasible in glioma patients. Clinical data were insufficient to draw conclusions regarding efficacy. Conclusions: KCRDs have positive effects on malignant gliomas in published preclinical studies. Preliminary clinical data suggest that KCRDs are safe and feasible. However, because of the paucity of clinical data, the efficacy of KCRDs for improving survival and quality of life of glioma patients remains to be proven in prospective studies.