Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Critical Reviews in Oncology / Hematology 153 (2020) 103061

Contents lists available at ScienceDirect

Critical Reviews in Oncology / Hematology


journal homepage: www.elsevier.com/locate/critrevonc

Review

Does nutrition for cancer patients feed the tumour? A clinical perspective T
a, b
F. Bozzetti *, Z. Stanga
a
Faculty of Medicine, University of Milan, Milan, Italy
b
Department of Diabetes, Endocrinology, Nutritional Medicine and Metabolism, Bern University Hospital and University of Bern, Bern, Switzerland

ARTICLE INFO ABSTRACT

Keywords: This review aims to answer to two basic questions: a) Which substrates does a tumour utilize and is there a
Nutrition and tumour growth regimen that might potentially favour the host over the tumour? and b) Does nutritional intervention dis-
Cancer cell metabolism proportionally affect tumour growth? Literature to date focuses on humans; although some references to mo-
Nutritive substrates of the cancer cell lecular mechanisms regulating cancer cells metabolism derive from studies on experimental tumours and cell
Nutrition of cancer patients
biology. Literature shows that some tumours, especially those of the brain and head/neck and lung, are glucose-
Calorie restriction
dependent, and patients with these tumours could benefit from a normocaloric ketogenic diet provided these
Ketogenic diet
Calorie restricted diet tumours exhibit high fluorodeoxyglucose (18F-FDG) captation. A high fat-protein, low carbohydrate diet appears
to better fulfil the nutritional requirements of the cancer patient. Current evidence shows no improvement in
tumoral response after restricting patients' caloric intake; whereas malnutrition is acknowledged as an important
negative predictive and prognostic factor in all cancer patients.

1. Introduction dietehave been transferred (perhaps inappropriately) to tumour-


bearing individuals, and some animal or cancer cell culture findings
The modern oncologistealso involved in coordinating nutritional have been prematurely adopted in clinical practice without considering
plans for his patientsefinds himself confronted with various conflicting the basic metabolic discrepancies between experimental and human
schools of thought which have increased in popularity over the last 20 tumours. It is well known, for instance, that the basal metabolic rate is
years. On the one hand, there is growing awareness of overfeeding, the 7-fold lower in humans than in rodents, and in advanced stage tumour
obesity epidemic in western countries, and its association with in- mass can account for up to 20 % of body weight. In humans, it rarely
creased rates of cancer, especially in young people. Several studies exceeds 0.1 % of the patient's weight. Such a difference could account
(mostly in animals, but also in overweight-obese humans) have shown for discrepancies in rodent and human response to dietary manipula-
the benefits of a calorie restricted diet (CRD) on some markers of tions such as CRD, ketogenic diets (KD) etc.
longevity and aging. The current research has alarmed some oncolo- Today's rapid data transmission may have caused information from
gists, as the administration of certain nutrients might be deleterious for epidemiology, basic research and clinical practice to overlap, creating
cancer patients, and hypocaloric diets have been marketed and widely an atmosphere of uncertainty or, even worse, of absolute fideism re-
advertised in the media. On the other hand, the clinical literature garding the benefits or risks of nutritional interventions in cancer pa-
clearly highlights the risk of short and long-term complications for tients.
malnourished patients undergoing oncologic treatment and its negative This appraisal of the literature focuses on clinical research or evi-
impact on quality of life and survival. The strategy of potentially re- dence limited to humans, and aims to answer two main questions:
versing malnutrition is also gaining popularity in clinical practice.
Nutritional interventions (via oral, enteral, and parenteral approaches) a Which substrates does the tumour utilize and, consequently, is there
have evolved tremendously thanks to the availability of nutritional a regimen which might potentially benefit the host and not the tu-
substrates, technological advances, and safety of administration since mour?
their introduction into clinical practice fifty years ago (Schwartz et al., b Does a nutritional intervention (normocaloric versus a CRD or spe-
1971). cial diet) disproportionally affect tumour growth?
The general impression is that some epidemiologic concepts of
cancer prevention in healthy peopleesuch as adopting a correct


Corresponding author at: Residenza Querce Milanodue, 20090, Segrate, Italy.
E-mail address: federicobozzetti@gmail.com (F. Bozzetti).

https://doi.org/10.1016/j.critrevonc.2020.103061
Received 10 March 2020; Received in revised form 6 July 2020; Accepted 7 July 2020
Available online 12 July 2020
1040-8428/ © 2020 Elsevier B.V. All rights reserved.
F. Bozzetti and Z. Stanga Critical Reviews in Oncology / Hematology 153 (2020) 103061

2. Biochemical characteristics and mutational alterations of NOTCH1 attenuate mitochondrial function, upregulate glycolysis, in-
cancer cell energy metabolism crease oxidative stress, or decrease oxidative phosphorylation. In ad-
dition, loss of function mutations/amplification of PIK3 and KRAS in-
2.1. Glucose dependency and its pathophysiologic meaning crease coupling glycolysis to the mitochondrial citric acid cycle, which
yields intermediates for the biosynthetic pathways, or decrease mi-
The idea that nutrition potentially stimulates tumour growth dates tochondrial respiration. In a recent study on human oral squamous
back to 1914. Rous, one century ago (Rous, 1914) showed that the cancer cell, Zheng (Zheng et al. (2019)) showed that STAT3 (signal
growth of transplantable tumour grafts could often be prevented or transducer and activator of transcription 3) promoted migration, in-
retarded by underfeeding the new host or by implementing a special vasion, epithelial-mesenchymal transition, as well as aerobic glycolysis
diet. These findings were subsequently replicated in other studies on of oral squamous cancer cells by inhibiting FOXO1 transcription. Si-
experimental brain (Mukherjee et al., 2004, 2002; Shelton et al., 2010; milar findings were reported in hepatocellular carcinoma (HCC) (Li
Yang et al., 2009), prostate, and breast tumours (Mukherjee et al., et al., 2017). According to Altenberg et al. (Altenberg and Greulich,
1999; De Lorenzo et al., 2011; Phoenix et al., 2010). 2004) glucose transporters and glycolytic enzymes are overexpressed in
Ten years later, Warburg (Warburg et al. (1927)) observed that 24 different types of cancer, including lung cancer.
normal cells produce energy in the mitochondria while cancer cells Finally, recent research has emphasized the role of long non-coding
produce energy in the cytosol, and hypothesized that mitochondrial RNAs (lncRNAs, a class of RNA molecules < 200 bp in length) in the
function was significantly impaired in cancer cells. Glycolysis, the regulation of cancer cell metabolism; primarily by regulating key en-
major pathway of glucose metabolism, takes place in the cytosol of all zymes in the glucose metabolism pathway (Fan et al., 2017).
cells and can occur aerobically or anaerobically depending on the Studies on HCC have shown that lncRNA Ftx and lncRNA SNHG3
availability of oxygen. This is metabolically significant as oxidation of interfere in glucose metabolism reprogramming through the PPAR-γ
glucose under aerobic conditions results in 32−36 mol of ATP/mol of pathway. Furthermore, they stimulate glycolysis by promoting the ex-
glucose, whereas only 2 mol of ATP can be produced under anaerobic pression and activity of phosphofructokinase and lactic-dehydrogenase,
conditions. In fact, the process of aerobic glycolysis occurs in two steps: while weakening the activity and expression of key tricarboxylic acid
the first occurs in the cytosol, involves the conversion of glucose to cycle enzymes such as isocitrate dehydrogenase and α-ketoglutarate
pyruvate, with resultant production of NADH and two molecules of dehydrogenase (Li et al., 2018).
ATP. Then, if oxygen is available, the free energy contained in NADH is For several decades researchers considered “anaerobic” glycolysis
further released via re-oxidization of the mitochondrial electron chain even in oxygen-rich conditions as simply filling a need for cancer cells
and results in the synthesis of 30 more mol of ATP/mol of glucose. living in a hypoxic environment and an inefficient way of procuring
When oxygen is in short supply, however, the NADH is re-oxidized by energy. The potential advantages of the Warburg effect for rapidly
reducing pyruvate to lactate through lactate dehydrogenase, and is growing cancer cells have only recently become apparent. They can be
secreted into the extracellular space via monocarboxylate transporters. summarised as follows (Cameron et al., 2018):
This severely limits the amount of ATP formed per mole of glucose
oxidized when compared with aerobic glycolysis.
Warburg (Warburg et al. (1927)) noted that shifting energy deri-
vation away from mitochondrial oxidative phosphorylation serves to
shuffle phosphometabolites into the pentose phosphate pathway for a) ATP is produced approximately 100 times faster than oxi-
biosynthesis of nucleic acids and lipids. The process of producing en- dative phosphorylation.
ergy mainly through non-oxidative breakdown of glucose obviously b) ROS generation is reduced, resulting in better preservation
requires an adequate availability of glucose - as glucose (not oxygen as of cancer cell genome.
in healthy human karyocytes) is used to produce ATP during anaerobic c) 5-phosphoribose-1-pyrophosphate is produced as an inter-
glycolysis. In cancer cells, the fermentation of glucose to lactate occurs mediate product of the pentose phosphate pathway, and is
even with sufficient oxygen supply, and lactate may reach concentra- used in the biosynthesis of purine and pyrimidine nucleo-
tions of up to 10−30 mM in cancer tissue that is 2–20 times higher than tides.
in healthy tissue (Brizel et al., 2001). This phenomenon is variously d) Lactic acid - for a long time only recognized as a “metabolic
termed “aerobic glycolysis”, “aerobic fermentation” (Fig. 1) (Pfeiffer waste product” - is now known to reduce cellular and tu-
and Morley, 2014). It was also observed that, for normal cells, the mour microenvironment pH (which ranges between
presence of glucose slightly increased respiration or had no effect on 6.0–6.5), as well as promote genetic changes which impair
oxygen consumption. On the contrary, glucose decreased oxygen up- anti-tumour immune response, reduce adherence cell junc-
take by cancer cells (so-called Crabtree effect). The Warburg effect in- tion, and facilitate detachment and metastases (Estrella
volves the alteration of metabolic enzymes, including hexokinase 2, et al., 2013; Garcia-Venzor et al., 2019). Lactate concentra-
pyruvate kinase type M2, glucose transporter 1, lactate dehydrogenase tion in whole breast tumour was found to correlate with
and lactate transporters (monocarboxilate transporters) (Fantin et al., Nottingham Prognostic Index in women with with invasive
2006; Mayer et al., 2014; Wang et al., 2014; Wong et al., 2015). ductal carcinoma (Cheung et al., 2020). Finally, recent re-
Notably, the Warburg phenotype has been associated not only with search reports that lactate is used continuously as a fuel
an energy gain, but also with the activation of numerous transcription under complete aerobic conditions by some tumours like
factors such as c-Myc, NF-kB, and Hypoxia-Inducible Factor 1-a (HIF 1- human lung cancer cells (Gallagher et al., 2009; Hirayama
a) (Fantin et al., 2006; Johnson and Perkins, 2012; Lu et al., 2002; Shim et al., 2009; Hui et al., 2017; Kennedy et al., 2013).
et al., 1997). These can affect the expression of metabolic enzymes, e) Abnormal metabolism may itself drive cancer progression
resulting in the deregulated conversion of glucose to lactate (Levine and through a preference for enzymatic isoforms that favour
Puzio-Kuter, 2010) which promotes a “tumour lactagenesis” state (San- anabolism (Ward and Thompson, 2012). For instance, pyr-
Millan and Brooks, 2017). Lactate can then be converted into pyruvate uvate kinase type M2 and hexokinase 2, key enzymes in the
which enters the TCA cycle in the mitochondria in the liver and forms cancer cell glycolytic pathway, seem to contribute to the
glucose (Cori cycle). This is subsequently used by cancer cells as a invasive potential and metastatic ability of the tumour
further energy source (Chen et al., 2016). (Anderson et al., 2017; Li et al., 2016).
There is evidence that genetic mutations can affect tumour cell
metabolism: loss of function mutations/deletions of PTEN, TP53,

2
F. Bozzetti and Z. Stanga Critical Reviews in Oncology / Hematology 153 (2020) 103061

Fig. 1. The Warburg effect.

2.2. Alternative energy sources and metabolic plasticity of the cancer cells 2011), while carbon and particularly nitrogen are used for biosynthesis.
This provides the substrate for purine and pyrimidine nucleotide
It is intriguing that human breast cancer cells with a Warburg synthesis (Mates et al., 2020, 2009) as well as a supply for amino groups
phenotype are able to evolve, survive, and proliferate independent of for non-essential amino acid synthesis. Especially in tumours which
glucose when they metastasize to brain. This is due to enhanced glu- exhibit low glycolytic activity, this cycle may present an alternative to
coneogenesis and oxidations of glutamine as well as branched chain glucose metabolism, or may simply provide an additional strategy for
amino acids, which together sustain the non-oxidative pentose pathway cancer cells to produce the energy necessary for continuous growth
for purine synthesis (Chen et al., 2015). (Lieberman et al., 2011).
Cancer cells that form dietary-resistant tumours carry mutations Under metabolic pressure, some tumour cells are able to take fatty
which cause constitutive activation of the PI3K pathway and, in culture, acids from their surroundings and perform β-oxidation to provide nu-
proliferate in the absence of insulin or IGF-1 (Kalaany and Sabatini, trients when glucose levels are low (Jeon et al., 2012). Many lncRNAs
2009). Approximately half of human melanomas harbour activating are involved in the regulation of lipid metabolism (Higashi et al., 2000;
mutations at amino acid V600 in the protein kinase BRAF, leading to Nguyen et al., 2007; van Solingen et al., 2018; Zeng et al., 2018). It is
constitutive activation of the mitogen-activated protein kinase (MAPK) noteworthy that the amount of ATP produced by the full oxidation of
pathway (Davies et al., 2002; Garnett and Marais, 2004). BRAF gene each fatty acid molecule is more than twice that of glucose (Boroughs
mutation occurs in 45–50 % of skin melanomas and the most frequent and DeBerardinis, 2015).
mutation involves the replacement of valine with glutamic acid at In conclusion, Warburg phenotype is found in many tumours (colon
codon 600 (V600E). BRAF inhibition decreases cellular glucose uptake cancer, liver metastases from colorectal cancer, squamous cell lung
in melanoma together with reduction in cell volume (Theodosakis et al., cancer, soft tissue sarcoma, glioblastoma, HCC, uterus carcinoma, head
2015). and neck cancer, and BRAF-negative melanoma (Brizel et al., 2001;
Some studies indicate that in certain tumours, glutamine transport Kennedy et al., 2013; Ward and Thompson, 2012; Wise et al., 2008;
and metabolism may be upregulated due to the presence of oncogenic Lieberman et al., 2011; Jeon et al., 2012; van Solingen et al., 2018;
levels of Myc, which induce a transcriptional programme promoting Zeng et al., 2018; Boroughs and DeBerardinis, 2015; Goodwin et al.,
glutaminolysis and trigger cellular to glutamine dependence as a 2017; Jiao et al., 2018; Liu et al., 2017; Marshall et al., 1979;
bioenergetic substrate (Wise et al., 2008). The utilization of glutamine Nikoobakht et al., 2019; Park et al., 2013; Schuurbiers et al., 2014;
within the mitochondria starts with its conversion to α-ketoglutarate Walenta et al., 2003, 1997; Walenta et al., 2000), and is absent in
mediated by glutaminase and glutamate dehydrogenase. Then α-ke- prostate adenocarcinoma and diffuse large B cell lymphoma, in 50 % of
toglutarate can be oxidized to succinate through α-ketoglutarate de- skin melanomas, 10 % of colorectal cancer, 100 % of hairy cell leu-
hydrogenase (the standard tricarboxylic acid cycle reaction) or alter- kaemia, 5% of multiple myeloma, and some thyroid cancers.
natively, can undergo reductive carboxylation by isocitrate
dehydrogenase to isocitrate and then to citrate (reverse tricarboxylic
acid cycle activity). Finally, the glutamine-derived citrate can be 3. Nutritional support and tumour growth in humans
transported to cytoplasm to generate acetyl-CoA for anabolic processes.
In conditions of hypoxic stress, glutamine-derived α-ketoglutarate is 3.1. Nutrient consumption by the human tumour in vivo
also used to stimulate lipid synthesis (Altman et al., 2016; Metallo et al.,
Three studies (Holm et al., 1995; Norton et al., 1980; van der Hulst

3
F. Bozzetti and Z. Stanga Critical Reviews in Oncology / Hematology 153 (2020) 103061

et al., 1997) measured blood flow using a non-invasive capacitance of GLUT1 whose expression is due to an oncogenic transcription effect
plethysmograph. They obtained arterial as well as venous effluent or to selective mutation. Furthermore, such effects may be lost during
blood samples from the tumour-bearing limb or intestinal segment, and the trajectory of the cancer cell towards metastatic colonization in
control was the healthy limb or the non-affected intestinal segment. The distant organs.
first study (Norton et al., 1980) showed a linear correlation between the
volume of soft tissue sarcoma of the limb and glucose uptake; which 3.2. Effects of the nutritional status, food intake or restriction on tumour
was approximately 1.3 g/g tumour/d in soft tissue sarcoma and about growth
0.3 g/g tumour/d in osteosarcoma. The study by Holm (Holm et al.,
1995) showed that the net uptake of glucose by the colonic tumour The presence of nutrients is associated with high levels of circu-
exceeded peripheral glucose uptake by a factor of 30 and lactate output lating glucose (and consequently of insulin), while the transcription of
from the tumour was 43 times higher than peripheral release. A net IGF-binding protein-1 decreases which in turn increases IGF-1 bioa-
release of glutamine occurred in 7 patients and net retention in 10. vailability (Rajaram et al., 1997). Through their binding to specific
Finally, the third study (van der Hulst et al., 1997) confirmed that colon tyrosine kinase receptors, insulin and free IGF-1 activate the phospha-
cancer had a higher glucose uptake and lactate release than healthy tidylinositol-3 kinase (PI3K)-Akt-mammalian target of rapamycin
intestine and showed that tumour-containing-colon did not extract complex 1 (mTORC1) pathways. TOR is a large (∼280 kDa) serine/
more glutamine than did non-tumour-containing colon. They also noted threonine protein kinase belonging to the PI3K-related kinase family. It
a higher tumoral uptake of essential amino acid and branched chain has also been shown that mTORC1 is regulated by growth factors
amino acid compared to a low peripheral release (Norton et al., 1980; through the phosphoinositide 3-kinase/protein kinase B, also known as
Hagmuller et al., 1995). Akt (PI3K/PKB or Akt) pathway, as well as the Ras/mitogen-activated
These studies and others using positron emission tomography with protein kinase (MAPK) pathway (Dibble and Cantley, 2015). mTORC1
fluorodeoxyglucose ((Lu et al., 2002)F-FDG) (Bozzetti et al., 2004; promotes proliferative signalling, increased cell proliferation, resistance
Nolop et al., 1987) attempted to quantify glucose uptake by the tumour to death, enhanced survival, and altered cellular metabolism - including
(glucose g per tumour 100 g/d) as well as tumour/healthy tissue up- increased fermentation of glucose and glutamine (Hanahan and
take. The approximate results were: colon cancer 8.2 (x4.2 tumour-free Weinberg, 2011).
colon), lung cancer 4.1 (x6.7 tumour-free lung), soft tissue sarcoma In comparison, dietary restriction activates the Nrf-2 gene (Lewis
(x1.8 tumour-free limb), liver metastases from colorectal cancer (x3.2 et al., 2010); an energy sensing network consisting of adenosine
tumour-free liver). monophosphate-activated protein kinase (AMPK), NAD-dependent
It was recently demonstrated that the facilitative glucose transporter deacetylase sirtuin-1 (SIRT1) (Sinclair, 2005) and SIRT 2 (Ahmed et al.,
1 (GLUT1) is responsible for augmented glucose uptake and metabolism 2019), peroxisome proliferator-activated receptor-α (PPAR-α) (Veech,
in several human cancers (Adekola et al., 2012). Various oncogenic 2004), and peroxisome - a proliferator-activated receptor-γ coactivator
transcription factors such as c-Myc may directly regulate GLUT1 mRNA 1-α (PGC-α) that counteracts the insulin/IGF-1PI3K-Akt-mTORC1
expression (Osthus et al., 2000), as well as the aberrant activation of pathway and promotes mitochondrial function (Cullingford, 2004). The
growth factor or oncogenic signalling pathways such as PI3K/AKT potential effects (metabolic reactions) of short-term fasting are depicted
(Wieman et al., 2007; Wofford et al., 2008). in Fig. 2. There is some epidemiologic evidence of slightly increased
Furthermore, microenvironment hypoxia also induces GLUT1 ex- risk of some cancers due to higher activity of the IGF system.
pression via the transcription factor, HIF-1α and the overexpression of In clinical practice, CRD is commonly defined as a long-term 20
lncRNA EIF3J-AS1 in hepatocellular carcinoma and lncRNA-HAL in %–40 % reduction of caloric intake through different forms of fasting.
breast cancer (Garcia-Venzor et al., 2019; Yang et al., 2019). A high These include intermittent fasting, alternate day fasting, two days a
expression of GLUT1 has been reported in oral squamous cell carcinoma week fasting, periodic fasting lasting three days or longer every two or
(Wang et al., 2018), non-small-cell lung carcinoma (NSCLC) (Chang more weeks, or simply maintaining a normal frequency of meals with
et al., 2019) and gastric cancer (Zhao et al., 2018). In osteosarcoma, reduced caloric content.
however, both GLUT1 and GLUT3 were highly expressed (Kang et al., Literature contains few scattered anecdotal reports showing benefits
2018), and in hepatocellular carcinomas GLUT1 and GLUT4 (Li et al., (Bell et al., 1964) or deleterious effects of enteral (EN) (English et al.,
2018; Wei et al., 2017) were found abnormally expressed. 1975) or parenteral (PN) (Rice and van Rij, 1987) feeding on tumour
Finally, Chen et al. (Chen et al., 2001) and Yun et al. (2009) have progression. Bozzetti et al. (Bozzetti et al., 1995) speculated that if
shown that the selective acquisition of KRAS or BRAF mutations in nutrients stimulate tumour growth, well-nourished patients should
response to glucose deprivation can upregulate GLUT1 expression. High harbour tumours with higher proliferation indexes as compared losing-
GLUT1 expression is clinically relevant to positron emission tomo- weight patients. However, in a sample of 136 patients with non-
graphy (PET) scanning, with the use of 18F-FDG (Nguyen et al., 2007) Hodgkin lymphoma they found an inverse statistically significant as-
for initial diagnosis and the prognostic evaluation of many tumours sociation between poor nutritional status and high labelling indexes.
including NSCLC (Li et al., 2018; Higashi et al., 2000; Wei et al., 2017; Malnourished patients had tumours with higher levels of incorporation
Mamede et al., 2005). of the 3H thymidine labelling index. Several studies (Champ et al.,
Recent studies (Nguyen et al., 2007; Boroughs and DeBerardinis, 2014; Fine et al., 2012; Zuccoli et al., 2010) investigated the potential
2015; Vesselle et al., 2008) have reported significant correlation be- role of CRD in very small series of patients undergoing hypocaloric
tween max-SUV and levels of GLUT1, between max-SUV and level of Ki- (sometimes ketogenic) diets of 9.3 (Zuccoli et al., 2010), 17 (Fine et al.,
67, and SUV and lung tumour growth (Duhaylongsod et al., 1995). 2012), 21 (Champ et al., 2014) kcal/kg/d for several weeks and only
Other noteworthy findings include that high plasma glucose levels reported the acceptable compliance of patients with such a regimen.
in patients with brain cancer is associated with shorter survival rates A 2009 literature review by Bozzetti and Mori (Bozzetti and Mori,
(Chaichana et al., 2010; Derr et al., 2009; McGirt et al., 2008); elevated 2009) (Table 1) investigating nutritional support and tumour growth in
SUV portends a poor prognosis in head and neck cancer patients humans found 12 studies (Baron et al., 1986; Bozzetti et al., 1999; Cao
(Kubicek et al., 2010) as well as those with epithelial ovarian cancer et al., 1994; Dionigi et al., 1991; Edstrom et al., 1989; Frank et al.,
(Chung et al., 2012); and, again, high GLUT1 expression is a predictor 1992; Jin et al., 1999; McNurlan et al., 1994; Ota et al., 1986; Pacelli
of distant recurrence and poor prognosis in rectal cancer patients after et al., 2007; Rossi-Fanelli et al., 1991; Westin et al., 1991) assessing
preoperative chemo-radiotherapy (Saigusa et al., 2012). tumour proliferation in connection with ornithine decarboxylase ac-
In conclusion, there is evidence that many (but not all) human tu- tivity, flow-cytometric DNA distribution, labelling index with tritiated
mours have a Warburg phenotype which is characterised by high levels thymidine or bromodeoxyuridine incorporation, DNA index, DNA

4
F. Bozzetti and Z. Stanga Critical Reviews in Oncology / Hematology 153 (2020) 103061

Fig. 2. Effects of starvation in a normal cell.


KB: ketone bodies; IGF-1: insulin-like growth
factor-1; IGFBP1: insulin-like growth factor-1
binding protein; inh IGFBP1: inhibitor of in-
sulin-like growth factor-1 binding protein; Akt:
protein-kinase B; Ras: rat sarcoma; PI3K:
phosphatidylinositol-3-kinase; Nrf: NF-kappaB
repressing factor; SIRT1: sirtulin1; FOXO:
forkhead box 0.

content and the percent of cells in S phase. phenotype may show higher tumour cell proliferation (not necessarily
Eight of the 12 studies reported that nutritional support appeared to tumour growth) in response to nutritional intervention and d) some
stimulate tumour cell proliferation. However, two considerations are discrepancies in results may be explained by the varying metabolic
noteworthy: first, tumour growth reflects the net balance of cell pro- tumour patterns in the different studies.
liferation and cell loss, and the above-mentioned indexes only explored
the first part of this ratio. Secondly, six of the eight studies reporting 3.3. Biologic and clinical effects of KD
tumour growth after nutritional support were in head and neck cancer
patients where tumour cell dependency on glucose is well known. More The KD highlights the interference of nutrients on tumour meta-
relevant information comes from two studies (Demark-Wahnefried bolism, by shifting from a quantitative number of calories to the qua-
et al., 2017; Henning et al., 2018) in overweight/obese patients with litative concept that glucose, regardless of the total calorie amount, is
prostate cancer who underwent a period of CRD before surgery. In the the preferred fuel for many cancers. However, the mechanism of action
first RCT (Demark-Wahnefried et al., 2017), 40 patients were rando- of KD is complex. Experimental studies (Stafford et al., 2010) on brain
mized either to receive their usual diet, or a regimen ranging from tumour indicate that KD effect on tumour growth is not simply due to a
16.6−14.5 kcal/kg/d during seven weeks to obtain weight loss of about reduction in available glucose, but is more likely to induce complex
4.7 kg. Contrary to the expectations (and compared with controls) the interactions amongst several gene networks that regulate important
weight-losing arm manifested significantly greater Ki67 proliferation intracellular signalling cascades and cellular homeostatic mechanisms.
rates at surgery. The second study (Henning et al., 2018) randomised 44 A recent review of this topic (Bozzetti and Zupec-Kania, 2016) lists
overweight/obese patients to hypocaloric regimen (minus three types of nutritional manipulation that produce the ketogenic
500−800 kcal/d from their usual diet) to achieve 3.7 kg loss after 5–8 state: a) total starvation without carbohydrate (CHO) or caloric intake,
weeks and found no significant changes in both malignant epithelium which is associated with the highest levels of ketones (KB) (3−7 mM),
apoptosis (TUNEL) or proliferation levels (Ki67). In keeping with these b) semi-starvation KD, similar to the protein-sparing regimen of <
biologic findings, a double blind RCT evaluating increases in nutrient 800 kcal/d, where CHO account for about 50 g daily and is associated
intake through administration of anamorelinea ghrelin analogue able with moderate levels of KB and finally c) mildly hypocaloric or nor-
to improve appetite in cachectic NSLC patientsereported a sharp in- mocaloric KD, providing variable amounts of protein, 70–80 % fat
crease in IGF-1 without any difference in survival at 4-month follow up calories, and usually more than 50 g CHO. These are associated with
of NSCLC patients compared to controls (Katakami et al., 2018; levels of KB of about 0.5−1 mM.
Takayama et al., 2016). A KD for cancer patients can be normocaloric, consisting of 70–80 %
In conclusion, the combined data would suggest a) that CRD does fat calories, no more than 50 g CHO and an adequate intake of high
not reduce tumour growth, as tumour proliferation is higher when quality proteins. As already mentioned, glucose dependency is due to
nutrient intake is reduced or nutritional status is compromised, b) in- several factors including activation of growth factor receptor/PI 3-ki-
creases in IGF-1 due to increased nutrient intake does not result in a nase/Akt signalling, loss of p53 wild-type activity and reduced ex-
poorer outcome, c) head and neck tumours exhibiting a Warburg pression of synthesis of cytochrome oxidase 2 necessary for

5
F. Bozzetti and Z. Stanga Critical Reviews in Oncology / Hematology 153 (2020) 103061

mitochondrial respiratory chain function, and loss of tp53-induced

No change in controls; significant ↑ in TPN; significant


No change in controls; significant ↑ in EN vs. controls
glycolysis and apoptosis regulator (TIGAR) which suppresses glycolysis.

No change in controls; no difference in intervention

No change or ↓ in controls; no change or ↑ in TPN


Finally, tumour hypoxia might stimulate accumulation of HIF-1α and
subsequent expression in the suppression of oxidative phosphorylation.

No change in controls; significant ↑ in TPN


A KD is inappropriate for cancer cells as these cells lack ketolytic en-
zymes, and β-hydroxybutyrate is an endogenous histone deacetylase
inhibitor which induces cell cycle arrest, differentiation and/or apop-
tosis and may improve mitochondrial function. Furthermore, KB has

No change in controls or TPN


also exhibited anticancer effects in several rodent tumour models.

Significant ↑ in IV patients
No change in both groups
Schroeder et al. (2013) investigated 12 patients with head and neck
↑ in TPN vs. controls
cancer. Using ultrasound, they implanted control small catheters inside
group vs. controls

vital tumour tissue or regional lymph node metastases, as well as in


Tumour growth

tumour-free submucosal tissue of the buccal region. After three days of


KD the mean lactate concentration declined to a greater extent in the
tumour tissue than in the tumour-free mucosa; demonstrating in vivo
the feasibility of interfering in tumour metabolism.
Bozzetti et al. (Bozzetti et al., 2004) studied 18F-FDG uptake in 12
Nutritional intervention (n, nutritional

35kcal and 1.4 g AA/kg as TPN

30kcal and 1.2 g AA/kg as TPN


42kcal/kg; 2.3 g AA/kg as TPN

25kcal/kg and 1.2 g AA/kg by


59kcal/kg and 1.9 g AA/kg as
Harris-Benedict x 1.2−1.5 as

Harris-Benedict x 1 and 1.5 g


Harris-Benedict x 1.5 as TPN

patients with metastatic liver from colorectal cancer who received


during the night alternatively a glucose-based or a lipid-based PN in-
fusion containing glucose 4 mg/kg/min or lipid 2 mg/kg/min, respec-
tively. Glucose captation by the metastases was 3.2 times higher than
AA/kg as TPN

by healthy liver tissue, increased very little after glucose PN and de-
creased very little after lipid PN. This was in contrast to captation by
IV route

the healthy liver, which increased 60 % with glucose PN and decreased


or EN

TPN

8% after lipid infusion. These findings suggested that metabolic activity


regimen)

EN

worked at a maximal regimen and was minimally affected (even if in


13

10
23

10

18

GI: gastrointestinal; AA: amino acid; iv: intravenous; EN: enteral nutrition; TPN: total parenteral nutrition; ↑: increase; ↓: decrease.

opposite directions) by glucose or lipid infusion.


9

Clinical application of KD was initially limited to a few case reports


19kcal; 1.1 g AA/kg by mouth or iv

Harris-Benedict x 1 and 1.5 g AA/


43kcal/kg; 1.4 g AA/kg by mouth

only assessing feasibility and tolerance of the regimen (Bozzetti et al.,


1996; Branca et al., 2015; Klement and Sweeney, 2016; Nebeling et al.,
Controls (n, usual hospital food)

1995). One interesting case was described by Seyfried et al. (Seyfried


Standard hospital food

et al., 2019) and Elsakka et al. (Elsakka et al., 2018) concerning glio-
blastoma: a patient who initially underwent an incomplete resection of
glioblastoma, followed by standard therapy with radiation and temo-
Regular food
Not reported

kg by mouth
< 1000kcal

zolomide, received KD for two years with progressive regression of the


Fasting

tumour, and died following complications from cerebral radionecrosis


without evidence of residual tumour. Another interesting case P. Kelly
(Children with Cancer UK, 2020) received, as the only treatment, KD
RCTs and comparative trials investigating nutritional support and tumour growth in humans.

13

23

10
7

for two years and four months after a diagnosis of inoperable glio-
blastoma, then had a probably complete tumour resection and is now
Malnourished, head and neck

still on KD and disease-free three years later.


Well-nourished, GI cancer

Well-nourished, GI cancer

More structured research is listed in Table 2 where only series of


Head and neck cancer

patients undergoing KD without oncologic treatment were included


(Bozzetti et al., 2004; Fine et al., 2012; Rossi-Fanelli et al., 1991;
Schroeder et al., 2013; Chu-Shore et al., 2010; Jansen and Walach,
2016; Rieger et al., 2014; Schmidt et al., 2011; Schwartz et al., 2015;
GI cancer

GI cancer
GI cancer

GI cancer
Primary

Tan-Shalaby et al., 2016). Ten studies (51 patients) evaluated the re-
cancer

sponse to KD, and stable disease or regression was reported in 20 (39


%). Although the potential for treating some patients with KD appears
Study duration (d)

promising, it should also be considered that at times study evaluation of


tumour response was suboptimal.
In conclusion, despite the presence of a biologic rationale for using a
Randomized clinical trials in malnourished patients

10−12
8−18

3−17

ketogenic regimen in patients with tumours showing a glucose de-


6−8

Comparative non- randomised clinical trials


10

15

pendency, clinical studies, albeit promising, are up to now confined to


7

small uncontrolled series with a short clinical follow up.


Rossi Fanelli (1991) [(Rossi-Fanelli et al.,
Edström (1989) [(Edstrom et al., 1989)

Bozzetti (1999) [(Bozzetti et al., 1999)


Dionigi (1991) [(Dionigi et al., 1991)

Pacelli (2007) [(Pacelli et al., 2007)

McNurlan (1994) [(McNurlan et al.,

3.4. Hypocaloric versus KD


Baron (1986) (Baron et al., 1986)
Jin (1999) [(Jin et al., 1999)

For reasons of exposition we dealt with CRD and KD separately,


however they share a common mechanism of action as they both rely
on increased body level of KB. Both CRD and KD lead to high rates of
fatty acid oxidation which increases production of acetyl-CoA. When
Author¨(year)

acetyl-CoA exceeds the capacity of tricarboxylic acid cycle to utilize it,


1991)

1994)

the production of β-hydroxybutyrate and acetoacetate, available as an


Table 1

energy source to the brain and other tissues (Cahill and Veech, 2003;
Morris, 2005; VanItallie and Nufert, 2003), increases. β-

6
F. Bozzetti and Z. Stanga Critical Reviews in Oncology / Hematology 153 (2020) 103061

hydroxybutyrate inhibits glycolysis (Cullingford, 2004) which explains

Non-significant ↓ in FDG uptake


why both fasting and KD can potentially improve antioxidative defense
= thymidine labelling index

Tumour mass ↓/= in 6/10


mechanisms in normal tissues (Veech, 2004; Shimazu et al., 2013;

= Tumour mass ↓ in 0/17


= Tumour mass in 7/11

Tumour mass ↓ in 3/11


Veech, 2006; Veech et al., 2001) but not in cancer cells. In humans, KB

Tumour mass ↓ in 0/2

Tumour mass ↓ in 7/7


Tumour lactate ↓ rise in the blood within 8−12 h after the start of fasting, reaching levels
= Tumour mass
up to 2−5 mM by 24 h (Cahill and Veech, 2003; Patel et al., 2019).
KB cannot be simply considered as fuel used during fasting as they
Outcome

are signalling molecules affecting cell and organ functions through the
expression of activity of several proteins and molecules, with capacity
to strongly influence metabolism (Newman and Verdin, 2017). These
include poly(adenosinediphosphate [ADP]–ribose) polymerase 1
(PARP1), and ADP ribosyl cyclase (CD38) (Lee, 2001), nicotinamide
adenine dinucleotide (NAD+), sirtuins (Imai and Guarente, 2016), the
< 70 g CHO (hypocaloric, normocaloric)
Fat vs. CHO, PN vs. oral diet (isocaloric)

PGC-1α, and the fibroblast growth factor 21 (Fisher and Maratos-Flier,


2016; Galman et al., 2008).
Cancer cells are unable to metabolize KB (Mates et al., 2020;
20−40 g CHO (normocaloric)
Fat vs. CHO PN (isocaloric)

Duhaylongsod et al., 1995; Maurer et al., 2011; Skinner et al., 2009)


Hypocaloric/normocaloric

due to mitochondrial defects (Wei et al., 2017; Zhou et al., 2007) and,
hence KB are toxic to some of them (Mates et al., 2020; Derr et al.,
2009; McGirt et al., 2008; Maurer et al., 2011; Skinner et al., 2009;
Non specified
Normocaloric
Normocaloric

Normocaloric

Magee et al., 1979; Scheck et al., 2010).


KD regimen

60 g CHO

KB could also act through an alteration of the expression of genes


involved in the cellular response to oxidative stress in tumour tissue -
notably COX-2, leading to a reduction in ROS (Chaichana et al., 2010;
Stafford et al., 2010). Cancer cells often have increased levels of ROS
(Fruehauf and Meyskens, 2007) which have been implicated in angio-
Colorectal cancer with liver metastases

genesis induction and tumour growth through the regulation of vas-


Head and neck squamous carcinoma

cular endothelial growth factor and HIF 1-α (Phoenix et al., 2010;
Weinberg and Chandel, 2009).
The rationale for adopting a CRD is based on preclinical data sug-
Tuberous sclerosis complex

gesting that the insulin pathway (including insulin, IGF-1 and the IGF-1
Miscellaneous cancers
Miscellaneous cancers

Miscellaneous cancers
Miscellaneous cancers

receptor) may be associated with cancer initiation and progression, and


that this pathway is upregulated through dietary consumption of CHO
and minimized by CHO restriction. Furthermore, cancer cells are unable
Glioblastoma
Glioblastoma

to use KB to their altered OXPHOS (Abdelwahab et al., 2012).


GI: gastrointestinal; CHO: carbohydrates; PN: parenteral nutrition; mths: months; ↑:increase; ↓:decrease.
GI cancer
Primary

Whereas the adoption of CRD in cancer patients undergoing onco-


logic treatment is at odds with the recommendations of both the
European Society for Clinical Nutrition and Metabolism (ESPEN)
(Arends et al., 2017) and American Society for Parenteral and Enteral
Nutrition (ASPEN) (August and Huhmann, 2009), some authors believe
Patients (n)

that the different utilization of KB by normal and cancer cell lines might
justify short-term fasting prior to chemotherapy. More precisely,
Effects of KD regimes on tumour outcome in patients without oncologic therapy.

27
12
12

10
16
17

14

healthy and tumour cells' contrasting reactions to nutrient scarcity is


4

2
7

termed "differential stress resistance" and may render tumour cells more
susceptible to the effects of chemotherapy; while at the same time
helping protect healthy cells against the toxic effects of chemotherapy
During infusion
Study duration

3 mths – 5 yrs

(Raffaghello et al., 2008). A recent RCT on 50 patients with glio-


3−12 mths

blastoma failed to find a benefit in survival for the arm receiving a CRD-
3 mths

5 mths
4 mths
6 wks

KG when compared to patients on a standard diet but an explorative


14 d

28 d
4d

analysis revealed that treated arm patients who had a glucose of less
than the median (83.5 mg/dl) on day 6 had a significantly longer sur-
vival compared to those above the median (Voss et al., 2020).
Without examining this emerging approach further, we'd like to cite
Rossi Fanelli (1991) [(Rossi-Fanelli et al., 1991)

Tan-Shalaby (2016) [(Tan-Shalaby et al., 2016)

the excellent reviews by de Cabo and Mattson (2019), and Shingler


Chu-Shore (2010) [(Chu-Shore et al., 2010)

Jansen (2016) [(Jansen and Walach, 2016)


Schroeder (2013) [(Schroeder et al., 2013)

et al. (Shingler et al., 2019) who collected 10 studies on KD, four studies
Schwartz (2015) [(Schwartz et al., 2015)
Schmidt (2013) [(Schmidt et al., 2011)
Bozzetti (2004) [(Bozzetti et al., 2004)

on fasting, and four combining some form of ketogenic or low CHO diet
Rieger (2014) [(Rieger et al., 2014)

with additional interventional aspects such as increased physical ac-


tivity or additional dietary changes.
Fine (2012) (Fine et al., 2012)

It is noteworthy that all seven KD studies which included KB levels


showed ketosis, or an increase in KB that was, however, not always
linked with a corresponding reduction in blood glucose. Meidenbauer
et al. and other suggest maintaining the ‘Glucose Ketone Index’ (the
Author (year)

molar ratio of circulating glucose over β-hydroxybutyrate)


(Meidenbauer et al., 2015) near one or below to achieve the highest
Table 2

benefit (Zuccoli et al., 2010; Rieger et al., 2014; Schwartz et al., 2015;
Winter et al., 2017).

7
F. Bozzetti and Z. Stanga Critical Reviews in Oncology / Hematology 153 (2020) 103061

The underlying hypothesis for a benefit in cancer is the reduction of If patients have a tumours with a high 18F-FDG captation, we might
signalling through insulin and growth hormone receptors, and an en- expect that such regimen maximally benefits the nutritional status of
hancement of FOXO and nuclear factor erythroid 2–related factor 2 the host in absence of a specific stimulation of the tumour growth. As
(Nrf2) transcription factors. However current evidence from interven- there is no correlation between blood glucose levels and 18 F-FDG
tional and dose escalation fasting studies does not show any reduction captation, there is no need for any correction in presence of hypergly-
in blood glucose (Dorff et al., 2016); with glucose even increasing after caemia (Eskian et al., 2019), even if for patients undergoing PN, it is
24 h in the RCT (de Groot et al., 2015). In addition, IGF analysis showed better withdrawing the infusion 4 h prior to the PET scan. A further test
lower than expected reductions in IGF-1 at 72 h (Dorff et al., 2016). to assess the glucose dependency of the tumour is the determination of
This finding is is in keeping with a recent meta-analysis which reported the transketolase‑like‑1 (TKTL1), a tumour-marking enzyme associated
that a CRD was not associated with a significant change in circulating with aerobic glycolysis of tumour cells which enhances the production
IGF-1 (Kazemi et al., 2020). It is interesting that short-term fasting of glucose-6-phosphate and glyceraldheide-3-phosphate. TKTL1 en-
contradicts all Societies of Anaesthesiology, which now suggest limiting zyme is a mutation of the transketolase gene, which seems to boost the
preoperative fasting as much as possible to avoid increases in insulin glucose metabolism of cancer cells (Coy et al., 2005; Langbein et al.,
resistance and the consequent anabolic resistance which is deleterious 2006; Sun et al., 2010) and parallels the progression of the tumour
for patient recovery. (Jansen and Walach, 2016). It can be reliably measured using an im-
Shingler et al. (Shingler et al., 2019) concluded that no re- munological assay, called the epitope detection in monocyte (EDIM)
commendation is possible at the moment and future research with test, with 94 % sensitivity and 82 % specificity.
adequately powered studies is required to test the effects of each dietary
restriction intervention on treatment toxicities and outcomes. 5. Conclusion
In conclusion, with reference to a hypothetical potentiation of
chemotherapy or a better compliance with its administration of KD or a In conclusion, it appears that below the seemingly simple question
CR regimen, clinical experience with variously combined KD or fasting whether feeding a cancer patient also feeds the tumour, there is a
regimens is rather scanty and did not provide any strong re- complex interplay of metabolic processes which also involve the acti-
commendation for the clinical practice. The preliminary findings of a vation of numerous transcription factors and the over- or under-
quite recent RCT on CRD was inconclusive and failed to demonstrate a expression of oncogenes. Whereas the exact scenario remains in a large
better tolerance of chemotherapy in patients undergoing CRD (de Groot part unknown, this review of the literature allows clarifying some main
et al., 2020). points:

4. Further considerations on the practical approach to feed the 1 Metabolic alterations (namely aerobic glycolysis) of the cancer cells
cancer patient does not simply represent an obligatory adaptation to a hostile en-
vironment, but may confer them an advantage in terms of growth
In the everyday practice, however, two other issues warrant con- and survival;
sideration to correctly plan a nutritional regimen to the cancer patient, 2 Cancer cells express a metabolic plasticity, which allows them to
namely the capability of substrate metabolic utilization of the tumour- overcome conditions of some substrates deprivation through the use
bearing patient and the effect of some nutrients on restoring the fat-free of alternative metabolic pathways;
mass of depleted patients. Various investigations on energy metabolism 3 Approximately 50 % of human tumours utilize glucose as preferred
in cancer patients (Li et al., 2017; Altenberg and Greulich, 2004; Fan fuel and this is the main rationale for CR or KD regimens;
et al., 2017; Li et al., 2018; Cameron et al., 2018; Arbeit et al., 1984; 4 CR does not reduce the tumour growth in humans as well as non-
Garcia-Canaveras et al., 2019; Hansell et al., 1986; Korber et al., 1999; restricted diet do not increase tumour growth;
Legaspi et al., 1987; Selberg et al., 1990; Shaw and Wolfe, 1987) have 5 The use of KD regimens maintains a strong nutritional rationale in
shown efficient mobilisation and oxidation of endogenous fat in the the cancer patient, with a good evidence as regards the effects on the
post-absorptive state, both in weight-stable and weight-losing cancer nutritional status of the patient and a little evidence regarding the
patients. Fat utilization ranged from 0.7 to 1.9 g/kg/d; that is about tumour growth control;
6.3−17 kcal/kg/d and approximately 60–78 % of the resting energy 6 There is no evidence, nowadays, of a strong clinical rationale for
expenditure of a patient. Similarly, another large study reported that using a calorie restricted diet in the malnourished cancer patient
cancer patients had a consistent non-protein respiratory quotient < 1, and that no study has yet determined whether intermittent fasting
indicating efficient use of fat substrates (Cao et al., 2010). Korber et al. affects cancer recurrence in humans (Nencioni et al., 2018).
(Korber et al., 1999) have shown improved clearance and oxidation of
lipid after administration of long-chain triglycerides or mixed long- Funding
chain/medium-chain triglycerides emulsions in cancer patients, as
compared with healthy controls. Metabolic and clinical experience with None declared.
high fat regimens are limited: Fearon et al. (Fearon et al., 1988) found
that KD maintained the nitrogen balance and whole-body protein Declaration of Competing Interest
synthesis, degradation, or turnover rates; Breitkreutz et al. (Breitkreutz
et al., 2005) showed in a RCT that malnourished patients on high-fat The authors have declared no conflicts of interest.
diet for 8 weeks gained weight and exhibited better fat-free and body
cell mass. They also reported enhanced quality of life compared with Acknowledgements
patients receiving standard nutrition. Cohen et al. (Cohen et al., 2018a,
b) recently found that overweight cancer patients on a hypocaloric KD The authors express their gratitude to Erica Holt for editing work
(∼5% of energy from CHO = 20 g/d, 25 % energy from protein and Emilie Reber for providing support for the preparation of the
=100 g/d, and 70 % energy from fat =125 g/d) lost total body fat manuscript.
while maintaining soft lean tissue and, compared with a standard diet,
showed improved physical function and increased energy. There is also References
evidence that such metabolic benefit may be potentiated by combining
nutrition with physical exercise and anti-inflammatory agents (Solheim Abdelwahab, M.G., Fenton, K.E., Preul, M.C., Rho, J.M., Lynch, A., Stafford, P., et al.,
et al., 2019). 2012. The ketogenic diet is an effective adjuvant to radiation therapy for the

8
F. Bozzetti and Z. Stanga Critical Reviews in Oncology / Hematology 153 (2020) 103061

treatment of malignant glioma. PLoS One 7 (5), e36197. 261–267. https://doi.org/10.1038/s41416-020-0886-7.


Adekola, K., Rosen, S.T., Shanmugam, M., 2012. Glucose transporters in cancer meta- Children with Cancer UK, 2020. Pablo Kelly. London (UK). Available from:. http://
bolism. Curr. Opin. Oncol. 24 (6), 650–654. www.childhoodcancer2018.org.uk/speakers/pablo-kelly.asp.
Ahmed, M.A., O’Callaghan, C., Chang, E.D., Jiang, H., Vassilopoulos, A., 2019. Context- Chung, H.H., Kwon, H.W., Kang, K.W., Park, N.H., Song, Y.S., Chung, J.K., et al., 2012.
dependent roles for SIRT2 and SIRT3 in tumor development upon calorie restriction Prognostic value of preoperative metabolic tumor volume and total lesion glycolysis
or high fat diet. Front. Oncol. 9, 1462. in patients with epithelial ovarian cancer. Ann. Surg. Oncol. 19 (6), 1966–1972.
Altenberg, B., Greulich, K.O., 2004. Genes of glycolysis are ubiquitously overexpressed in Chu-Shore, C.J., Major, P., Camposano, S., Muzykewicz, D., Thiele, E.A., 2010. The
24 cancer classes. Genomics 84 (6), 1014–1020. natural history of epilepsy in tuberous sclerosis complex. Epilepsia 51 (7),
Altman, B.J., Stine, Z.E., Dang, C.V., 2016. From Krebs to clinic: glutamine metabolism to 1236–1241.
cancer therapy. Nat. Rev. Cancer 16 (10), 619–634. Cohen, C.W., Fontaine, K.R., Arend, R.C., Alvarez, R.D., Leath III, C.A., Huh, W.K., et al.,
Anderson, M., Marayati, R., Moffitt, R., Yeh, J.J., 2017. Hexokinase 2 promotes tumor 2018a. A ketogenic diet reduces central obesity and serum insulin in women with
growth and metastasis by regulating lactate production in pancreatic cancer. ovarian or endometrial cancer. J. Nutr. 148 (8), 1253–1260.
Oncotarget 8 (34), 56081–56094. Cohen, C.W., Fontaine, K.R., Arend, R.C., Soleymani, T., Gower, B.A., 2018b. Favorable
Arbeit, J.M., Lees, D.E., Corsey, R., Brennan, M.F., 1984. Resting energy expenditure in effects of a ketogenic diet on physical function, perceived energy, and food cravings
controls and cancer patients with localized and diffuse disease. Ann. Surg. 199 (3), in women with ovarian or endometrial cancer: a randomized, controlled trial.
292–298. Nutrients 10 (9).
Arends, J., Bachmann, P., Baracos, V., Barthelemy, N., Bertz, H., Bozzetti, F., et al., 2017. Coy, J.F., Dressler, D., Wilde, J., Schubert, P., 2005. Mutations in the transketolase-like
ESPEN guidelines on nutrition in cancer patients. Clin. Nutr. (Edinburgh, Scotland). gene TKTL1: clinical implications for neurodegenerative diseases, diabetes and
36 (1), 11–48. cancer. Clin. Lab. 51 (5-6), 257–273.
August, D.A., Huhmann, M.B., 2009. A.S.P.E.N. Clinical guidelines: nutrition support Cullingford, T.E., 2004. The ketogenic diet; fatty acids, fatty acid-activated receptors and
therapy during adult anticancer treatment and in hematopoietic cell transplantation. neurological disorders. Prostaglandins Leukot. Essent. Fatty Acids 70 (3), 253–264.
South Afr. J. Clin. Nutr. 33 (5), 472–500. Davies, H., Bignell, G.R., Cox, C., Stephens, P., Edkins, S., Clegg, S., et al., 2002. Mutations
Baron, P.L., Lawrence Jr, W., Chan, W.M., White, F.K., Banks Jr, W.L., 1986. Effects of of the BRAF gene in human cancer. Nature 417 (6892), 949–954.
parenteral nutrition on cell cycle kinetics of head and neck cancer. Arch. Surg. de Cabo, R., Mattson, M.P., 2019. Effects of intermittent fasting on health, aging, and
(Chicago, Ill : 1960) 121 (11), 1282–1286. disease. N. Engl. J. Med. 381 (26), 2541–2551.
Bell, J.W., Jesseph, J.E., Leighton, R.S., 1964. Spontaneous regression of bronchogenic de Groot, S., Vreeswijk, M.P., Welters, M.J., Gravesteijn, G., Boei, J.J., Jochems, A., et al.,
carcinoma with five year survival. J. Thorac. Cardiovasc. Surg. 48, 984–990. 2015. The effects of short-term fasting on tolerance to (neo) adjuvant chemotherapy
Boroughs, L.K., DeBerardinis, R.J., 2015. Metabolic pathways promoting cancer cell in HER2-negative breast cancer patients: a randomized pilot study. BMC Cancer 15,
survival and growth. Nat. Cell Biol. 17 (4), 351–359. 652.
Bozzetti, F., Boracchi, P., Costa, A., Cozzaglio, L., Battista, A., Giori, A., et al., 1995. de Groot, S., Lugtenberg, R.T., Cohen, D., et al., 2020. Fasting mimicking diet as an ad-
Relationship between nutritional status and tumor growth in humans. Tumori 81 junct to neoadjuvant chemotherapy for breast cancer in the multicentre randomized
(1), 1–6. phase 2 DIRECT trial. Nat Commun. 11 (1), 3083. https://doi.org/10.1038/s41467-
Bozzetti, F., Cozzaglio, L., Gavazzi, C., Brandi, S., Bonfanti, G., Lattarulo, M., et al., 1996. 020-16138-3. Published 2020 Jun 23.
Total nutritional manipulation in humans: report of a cancer patient. Clin. Nutr. De Lorenzo, M.S., Baljinnyam, E., Vatner, D.E., Abarzua, P., Vatner, S.F., Rabson, A.B.,
(Edinburgh, Scotland) 15 (4), 207–209. 2011. Caloric restriction reduces growth of mammary tumors and metastases.
Bozzetti, F., Gavazzi, C., Cozzaglio, L., Costa, A., Spinelli, P., Viola, G., 1999. Total par- Carcinogenesis 32 (9), 1381–1387.
enteral nutrition and tumor growth in malnourished patients with gastric cancer. Demark-Wahnefried, W., Rais-Bahrami, S., Desmond, R.A., Gordetsky, J.B., Hunter, G.R.,
Tumori 85 (3), 163–166. Yang, E.S., et al., 2017. Presurgical weight loss affects tumour traits and circulating
Bozzetti, F., Gavazzi, C., Mariani, L., Crippa, F., 2004. Glucose-based total parenteral biomarkers in men with prostate cancer. Br. J. Cancer 117 (9), 1303–1313.
nutrition does not stimulate glucose uptake by humans tumours. Clin. Nutr. Derr, R.L., Ye, X., Islas, M.U., Desideri, S., Saudek, C.D., Grossman, S.A., 2009.
(Edinburgh, Scotland) 23 (3), 417–421. Association between hyperglycemia and survival in patients with newly diagnosed
Bozzetti, F., Mori, V., 2009. Nutritional support and tumour growth in humans: a nar- glioblastoma. J. Clin. Oncol. 27 (7), 1082–1086.
rative review of the literature. Clin Nutr. 28 (3), 226–230. https://doi.org/10.1016/j. Dibble, C.C., Cantley, L.C., 2015. Regulation of mTORC1 by PI3K signaling. Trends Cell
clnu.2009.02.006. Biol. 25 (9), 545–555.
Bozzetti, F., Zupec-Kania, B., 2016. Toward a cancer-specific diet. Clin. Nutr. (Edinburgh, Dionigi, P., Jemos, V., Cebrelli, T., Ferrari, C., Ferrari, A., Berizzi, F., et al., 1991. Pre-
Scotland) 35 (5), 1188–1195. operative nutritional support and tumour cell kinetics in malnourished patients with
Branca, Jj, Pacini, S., Ruggiero, M., 2015. Effects of pre-surgical vitamin D supple- gastric cancer. Clin. Nutr. (Edinburgh, Scotland) 10 (Suppl), 77–84.
mentation and ketogenic diet in a patient with recurrent breast cancer. Anticancer Dorff, T.B., Groshen, S., Garcia, A., Shah, M., Tsao-Wei, D., Pham, H., et al., 2016. Safety
Res. 35 (10), 5525–5532. and feasibility of fasting in combination with platinum-based chemotherapy. BMC
Breitkreutz, R., Tesdal, K., Jentschura, D., Haas, O., Leweling, H., Holm, E., 2005. Effects Cancer 16, 360.
of a high-fat diet on body composition in cancer patients receiving chemotherapy: a Duhaylongsod, F.G., Lowe, V.J., Patz Jr, E.F., Vaughn, A.L., Coleman, R.E., Wolfe, W.G.,
randomized controlled study. Wien. Klin. Wochenschr. 117 (19-20), 685–692. 1995. Lung tumor growth correlates with glucose metabolism measured by fluoride-
Brizel, D.M., Schroeder, T., Scher, R.L., Walenta, S., Clough, R.W., Dewhirst, M.W., et al., 18 fluorodeoxyglucose positron emission tomography. Ann. Thorac. Surg. 60 (5),
2001. Elevated tumor lactate concentrations predict for an increased risk of metas- 1348–1352.
tases in head-and-neck cancer. Int. J. Radiat. Oncol. Biol. Phys. 51 (2), 349–353. Edstrom, S., Westin, T., Delle, U., Lundholm, K., 1989. Cell cycle distribution and or-
Cahill Jr, G.F., Veech, R.L., 2003. Ketoacids? Good medicine? Trans. Am. Clin. Climatol. nithine decarboxylase activity in head and neck cancer in response to enteral nutri-
Assoc. 114, 149–161 discussion 62-63. tion. Eur. J. Cancer Clin. Oncol. 25 (2), 227–232.
Cameron, M.E., Yakovenko, A., Trevino, J.G., 2018. Glucose and lactate transport in Elsakka, Ama, Bary, Ma, Abdelzaher, E., Elnaggar, M., Kalamian, M., Mukherjee, P., et al.,
pancreatic cancer: glycolytic metabolism revisited. J. Oncol. 2018, 6214838. 2018. Management of glioblastoma multiforme in a patient treated with ketogenic
Cao, W.X., Xiao, H.B., Yin, H.R., 1994. [Effects of preoperative parenteral nutritional metabolic therapy and modified standard of care: a 24-month follow-up. Front. Nutr.
support with chemotherapy on tumor cell kinetics in gastric cancer patients]. 5, 20.
Zhonghua zhong liu za zhi [Chin. J. Oncol.] 16 (2), 137–140. English 2nd, Wj, Suskind, R., Damrongsak, D., Kulapongs, P., Olson, Re., 1975. Can the
Cao, D.X., Wu, G.H., Zhang, B., Quan, Y.J., Wei, J., Jin, H., et al., 2010. Resting energy growth of a neuroblastoma be influenced by a child’s nutritional state? Observations
expenditure and body composition in patients with newly detected cancer. Clin. Nutr. in a patient treated for kwashiorkor and later given a restricted diet. Clin. Pediatr.
(Edinburgh, Scotland) 29 (1), 72–77. (Phila) 14 (9), 868–869.
Chaichana, K.L., McGirt, M.J., Woodworth, G.F., Datoo, G., Tamargo, R.J., Weingart, J., Eskian, M., Alavi, A., Khorasanizadeh, M., Viglianti, B.L., Jacobsson, H., Barwick, T.D.,
et al., 2010. Persistent outpatient hyperglycemia is independently associated with et al., 2019. Effect of blood glucose level on standardized uptake value (SUV) in (18)
survival, recurrence and malignant degeneration following surgery for hemispheric F- FDG PET-scan: a systematic review and meta-analysis of 20,807 individual SUV
low grade gliomas. Neurol. Res. 32 (4), 442–448. measurements. Eur. J. Nucl. Med. Mol. Imaging 46 (1), 224–237.
Champ, C.E., Palmer, J.D., Volek, J.S., Werner-Wasik, M., Andrews, D.W., Evans, J.J., Estrella, V., Chen, T., Lloyd, M., Wojtkowiak, J., Cornnell, H.H., Ibrahim-Hashim, A.,
et al., 2014. Targeting metabolism with a ketogenic diet during the treatment of et al., 2013. Acidity generated by the tumor microenvironment drives local invasion.
glioblastoma multiforme. J. Neurooncol. 117 (1), 125–131. Cancer Res. 73 (5), 1524–1535.
Chang, L., Xu, W., Zhang, Y., Gong, F., 2019. Long non-coding RNA-NEF targets glucose Fan, C., Tang, Y., Wang, J., Xiong, F., Guo, C., Wang, Y., et al., 2017. Role of long non-
transportation to inhibit the proliferation of non-small-cell lung cancer cells. Oncol. coding RNAs in glucose metabolism in cancer. Mol. Cancer 16 (1), 130.
Lett. 17 (3), 2795–2801. Fantin, V.R., St-Pierre, J., Leder, P., 2006. Attenuation of LDH-A expression uncovers a
Chen, C., Pore, N., Behrooz, A., Ismail-Beigi, F., Maity, A., 2001. Regulation of glut1 link between glycolysis, mitochondrial physiology, and tumor maintenance. Cancer
mRNA by hypoxia-inducible factor-1. Interaction between H-ras and hypoxia. J. Biol. Cell 9 (6), 425–434.
Chem. 276 (12), 9519–9525. Fearon, K.C., Borland, W., Preston, T., Tisdale, M.J., Shenkin, A., Calman, K.C., 1988.
Chen, J., Lee, H.J., Wu, X., Huo, L., Kim, S.J., Xu, L., et al., 2015. Gain of glucose-in- Cancer cachexia: influence of systemic ketosis on substrate levels and nitrogen me-
dependent growth upon metastasis of breast cancer cells to the brain. Cancer Res. 75 tabolism. Am. J. Clin. Nutr. 47 (1), 42–48.
(3), 554–565. Fine, E.J., Segal-Isaacson, C.J., Feinman, R.D., Herszkopf, S., Romano, M.C., Tomuta, N.,
Chen, Y.J., Mahieu, N.G., Huang, X., Singh, M., Crawford, P.A., Johnson, S.L., et al., 2016. et al., 2012. Targeting insulin inhibition as a metabolic therapy in advanced cancer: a
Lactate metabolism is associated with mammalian mitochondria. Nat. Chem. Biol. 12 pilot safety and feasibility dietary trial in 10 patients. Nutrition (Burbank, Los
(11), 937–943. Angeles County, Calif). 28 (10), 1028–1035.
Cheung, S.M., Husain, E., Masannat, Y., et al., 2020. Lactate concentration in breast Fisher, F.M., Maratos-Flier, E., 2016. Understanding the physiology of FGF21. Annu. Rev.
cancer using advanced magnetic resonance spectroscopy. Br J Cancer. 123 (2), Physiol. 78, 223–241.

9
F. Bozzetti and Z. Stanga Critical Reviews in Oncology / Hematology 153 (2020) 103061

Frank, J.L., Lawrence Jr, W., Banks Jr, W.L., McKinnon, J.G., Chan, W.M., Collins, J.M., Langbein, S., Zerilli, M., Zur Hausen, A., Staiger, W., Rensch-Boschert, K., Lukan, N.,
1992. Modulation of cell cycle kinetics in human cancer with total parenteral nu- et al., 2006. Expression of transketolase TKTL1 predicts colon and urothelial cancer
trition. Cancer 69 (7), 1858–1864. patient survival: warburg effect reinterpreted. Br. J. Cancer 94 (4), 578–585.
Fruehauf, J.P., Meyskens Jr, F.L., 2007. Reactive oxygen species: a breath of life or death? Lee, H.C., 2001. Physiological functions of cyclic ADP-ribose and NAADP as calcium
Clin. Cancer Res. 13 (3), 789–794. messengers. Annu. Rev. Pharmacol. Toxicol. 41, 317–345.
Gallagher, C.N., Carpenter, K.L., Grice, P., Howe, D.J., Mason, A., Timofeev, I., et al., Legaspi, A., Jeevanandam, M., Starnes Jr, H.F., Brennan, M.F., 1987. Whole body lipid
2009. The human brain utilizes lactate via the tricarboxylic acid cycle: a 13C-labelled and energy metabolism in the cancer patient. Metab. Clin. Exp. 36 (10), 958–963.
microdialysis and high-resolution nuclear magnetic resonance study. Brain 132 (Pt Levine, A.J., Puzio-Kuter, A.M., 2010. The control of the metabolic switch in cancers by
10), 2839–2849. oncogenes and tumor suppressor genes. Science (New York, NY). 330 (6009),
Galman, C., Lundasen, T., Kharitonenkov, A., Bina, H.A., Eriksson, M., Hafstrom, I., et al., 1340–1344.
2008. The circulating metabolic regulator FGF21 is induced by prolonged fasting and Lewis, K.N., Mele, J., Hayes, J.D., Buffenstein, R., 2010. Nrf2, a guardian of healthspan
PPARalpha activation in man. Cell Metab. 8 (2), 169–174. and gatekeeper of species longevity. Integr. Comp. Biol. 50 (5), 829–843.
Garcia-Canaveras, J.C., Chen, L., Rabinowitz, J.D., 2019. The tumor metabolic micro- Li, C., Zhao, Z., Zhou, Z., Liu, R., 2016. PKM2 promotes cell survival and invasion under
environment: lessons from lactate. Cancer Res. 79 (13), 3155–3162. metabolic. stress by enhancing Warburg effect in pancreatic ductal adenocarcinoma.
Garcia-Venzor, A., Mandujano-Tinoco, E.A., Lizarraga, F., Zampedri, C., Krotzsch, E., Dig. Dis. Sci. 61 (3), 767–773.
Salgado, R.M., et al., 2019. Microenvironment-regulated lncRNA-HAL is able to Li, M., Jin, R., Wang, W., Zhang, T., Sang, J., Li, N., et al., 2017. STAT3 regulates gly-
promote stemness in breast cancer cells. Biochim. Biophys. Acta Mol. Cell Res. 1866 colysis via targeting hexokinase 2 in hepatocellular carcinoma cells. Oncotarget 8
(12), 118523. (15), 24777–24784.
Garnett, M.J., Marais, R., 2004. Guilty as charged: B-RAF is a human oncogene. Cancer Li, X., Zhao, Q., Qi, J., Wang, W., Zhang, D., Li, Z., et al., 2018. lncRNA Ftx promotes
Cell 6 (4), 313–319. aerobic glycolysis and tumor progression through the PPARgamma pathway in he-
Goodwin, J., Neugent, M.L., Lee, S.Y., Choe, J.H., Choi, H., Jenkins, D.M.R., et al., 2017. patocellular carcinoma. Int. J. Oncol. 53 (2), 551–566.
The distinct metabolic phenotype of lung squamous cell carcinoma defines selective Lieberman, B.P., Ploessl, K., Wang, L., Qu, W., Zha, Z., Wise, D.R., et al., 2011. PET
vulnerability to glycolytic inhibition. Nat. Commun. 8, 15503. imaging of glutaminolysis in tumors by 18F-(2S,4R)4-fluoroglutamine. J. Nucl. Med.
Hagmuller, E., Kollmar, H.B., Gunther, H.J., Holm, E., Trede, M., 1995. Protein meta- 52 (12), 1947–1955.
bolism in human colon carcinomas: in vivo investigations using a modified tracer Liu, Y., Murray-Stewart, T., Casero Jr, R.A., Kagiampakis, I., Jin, L., Zhang, J., et al., 2017.
technique with L-[1-13C]leucine. Cancer Res. 55 (5), 1160–1167. Targeting hexokinase 2 inhibition promotes radiosensitization in HPV16 E7-induced
Hanahan, D., Weinberg, R.A., 2011. Hallmarks of cancer: the next generation. Cell 144 cervical cancer and suppresses tumor growth. Int. J. Oncol. 50 (6), 2011–2023.
(5), 646–674. Lu, H., Forbes, R.A., Verma, A., 2002. Hypoxia-inducible factor 1 activation by aerobic
Hansell, D.T., Davies, J.W., Shenkin, A., Burns, H.J., 1986. The oxidation of body fuel glycolysis implicates the Warburg effect in carcinogenesis. J. Biol. Chem. 277 (26),
stores in cancer patients. Ann. Surg. 204 (6), 637–642. 23111–23115.
Henning, S.M., Galet, C., Gollapudi, K., Byrd, J.B., Liang, P., Li, Z., et al., 2018. Phase II Magee, B.A., Potezny, N., Rofe, A.M., Conyers, R.A., 1979. The inhibition of malignant
prospective randomized trial of weight loss prior to radical prostatectomy. Prostate cell growth by ketone bodies. Aust. J. Exp. Biol. Med. Sci. 57 (5), 529–539.
Cancer Prostatic Dis. 21 (2), 212–220. Mamede, M., Higashi, T., Kitaichi, M., Ishizu, K., Ishimori, T., Nakamoto, Y., et al., 2005.
Higashi, K., Ueda, Y., Sakurai, A., Mingwang, X., Xu, L., Murakami, M., et al., 2000. [18F]FDG uptake and PCNA, Glut-1, and Hexokinase-II expressions in cancers and
Correlation of Glut-1 glucose transporter expression with [(18)F]FDG uptake in non- inflammatory lesions of the lung. Neoplasia (New York, NY). 7 (4), 369–379.
small cell lung cancer. Eur. J. Nucl. Med. 27 (12), 1778–1785. Marshall, M.J., Neal, F.E., Goldberg, D.M., 1979. Isoenzymes of hexokinase, 6-phos-
Hirayama, A., Kami, K., Sugimoto, M., Sugawara, M., Toki, N., Onozuka, H., et al., 2009. phogluconate dehydrogenase, phosphoglucomutase and lactate dehydrogenase in
Quantitative metabolome profiling of colon and stomach cancer microenvironment uterine cancer. Br. J. Cancer 40 (3), 380–390.
by capillary electrophoresis time-of-flight mass spectrometry. Cancer Res. 69 (11), Mates, J.M., Segura, J.A., Campos-Sandoval, J.A., Lobo, C., Alonso, L., Alonso, F.J., et al.,
4918–4925. 2009. Glutamine homeostasis and mitochondrial dynamics. Int. J. Biochem. Cell Biol.
Holm, E., Hagmuller, E., Staedt, U., Schlickeiser, G., Gunther, H.J., Leweling, H., et al., 41 (10), 2051–2061.
1995. Substrate balances across colonic carcinomas in humans. Cancer Res. 55 (6), Mates, J.M., Di Paola, F.J., Campos-Sandoval, J.A., Mazurek, S., Marquez, J., 2020.
1373–1378. Therapeutic targeting of glutaminolysis as an essential strategy to combat cancer.
Hui, S., Ghergurovich, J.M., Morscher, R.J., Jang, C., Teng, X., Lu, W., et al., 2017. Semin. Cell Dev. Biol. 98, 34–43.
Glucose feeds the TCA cycle via circulating lactate. Nature 551 (7678), 115–118. Maurer, G.D., Brucker, D.P., Bahr, O., Harter, P.N., Hattingen, E., Walenta, S., et al., 2011.
Imai, S.I., Guarente, L., 2016. It takes two to tango: NAD(+) and sirtuins in aging/ Differential utilization of ketone bodies by neurons and glioma cell lines: a rationale
longevity control. NPJ Aging Mech. Dis. 2, 16017. for ketogenic diet as experimental glioma therapy. BMC Cancer 11, 315.
Jansen, N., Walach, H., 2016. The development of tumours under a ketogenic diet in Mayer, A., Schmidt, M., Seeger, A., Serras, A.F., Vaupel, P., Schmidberger, H., 2014.
association with the novel tumour marker TKTL1: a case series in general practice. GLUT-1 expression is largely unrelated to both hypoxia and the Warburg phenotype
Oncol. Lett. 11 (1), 584–592. in squamous cell carcinomas of the vulva. BMC Cancer 14, 760.
Jeon, S.M., Chandel, N.S., Hay, N., 2012. AMPK regulates NADPH homeostasis to pro- McGirt, M.J., Chaichana, K.L., Gathinji, M., Attenello, F., Than, K., Ruiz, A.J., et al., 2008.
mote tumour cell survival during energy stress. Nature 485 (7400), 661–665. Persistent outpatient hyperglycemia is independently associated with decreased
Jiao, L., Zhang, H.L., Li, D.D., Yang, K.L., Tang, J., Li, X., et al., 2018. Regulation of survival after primary resection of malignant brain astrocytomas. Neurosurgery 63
glycolytic metabolism by autophagy in liver cancer involves selective autophagic (2), 286–291 discussion 91.
degradation of HK2 (hexokinase 2). Autophagy 14 (4), 671–684. McNurlan, M.A., Heys, S.D., Park, K.G., Broom, J., Brown, D.S., Eremin, O., et al., 1994.
Jin, D., Phillips, M., Byles, J.E., 1999. Effects of parenteral nutrition support and che- Tumour and host tissue responses to branched-chain amino acid supplementation of
motherapy on the phasic composition of tumor cells in gastrointestinal cancer. South patients with cancer. Clin. Sci. 86 (3), 339–345.
Afr. J. Clin. Nutr. 23 (4), 237–241. Meidenbauer, J.J., Mukherjee, P., Seyfried, T.N., 2015. The glucose ketone index calcu-
Johnson, R.F., Perkins, N.D., 2012. Nuclear factor-kappaB, p53, and mitochondria: reg- lator: a simple tool to monitor therapeutic efficacy for metabolic management of
ulation of cellular metabolism and the Warburg effect. Trends Biochem. Sci. 37 (8), brain cancer. Nutr Metab. 12, 12.
317–324. Metallo, C.M., Gameiro, P.A., Bell, E.L., Mattaini, K.R., Yang, J., Hiller, K., et al., 2011.
Kalaany, N.Y., Sabatini, D.M., 2009. Tumours with PI3K activation are resistant to dietary Reductive glutamine metabolism by IDH1 mediates lipogenesis under hypoxia.
restriction. Nature 458 (7239), 725–731. Nature 481 (7381), 380–384.
Kang, Y., Zhu, X., Xu, Y., Tang, Q., Huang, Z., Zhao, Z., et al., 2018. Energy stress-induced Morris, A.A., 2005. Cerebral ketone body metabolism. J. Inherit. Metab. Dis. 28 (2),
lncRNA HAND2-AS1 represses HIF1alpha-mediated energy metabolism and inhibits 109–121.
osteosarcoma progression. Am. J. Cancer Res. 8 (3), 526–537. Mukherjee, P., Sotnikov, A.V., Mangian, H.J., Zhou, J.R., Visek, W.J., Clinton, S.K., 1999.
Katakami, N., Uchino, J., Yokoyama, T., Naito, T., Kondo, M., Yamada, K., et al., 2018. Energy intake and prostate tumor growth, angiogenesis, and vascular endothelial
Anamorelin (ONO-7643) for the treatment of patients with non-small cell lung cancer growth factor expression. J. Natl. Cancer Inst. 91 (6), 512–523.
and cachexia: results from a randomized, double-blind, placebo-controlled, multi- Mukherjee, P., El-Abbadi, M.M., Kasperzyk, J.L., Ranes, M.K., Seyfried, T.N., 2002.
center study of Japanese patients (ONO-7643-04). Cancer 124 (3), 606–616. Dietary restriction reduces angiogenesis and growth in an orthotopic mouse brain
Kennedy, K.M., Scarbrough, P.M., Ribeiro, A., Richardson, R., Yuan, H., Sonveaux, P., tumour model. Br. J. Cancer 86 (10), 1615–1621.
et al., 2013. Catabolism of exogenous lactate reveals it as a legitimate metabolic Mukherjee, P., Abate, L.E., Seyfried, T.N., 2004. Antiangiogenic and proapoptotic effects
substrate in breast cancer. PLoS One 8 (9), e75154. of dietary restriction on experimental mouse and human brain tumors. Clin. Cancer
Kazemi, A., Speakman, J.R., Soltani, S., Djafarian, K., 2020. Effect of calorie restriction or Res. 10 (16), 5622–5629.
protein intake on circulating levels of insulin like growth factor I in humans: A sys- Nebeling, L.C., Miraldi, F., Shurin, S.B., Lerner, E., 1995. Effects of a ketogenic diet on
tematic review and meta-analysis. Clin Nutr. 39 (6), 1705–1716. https://doi.org/10. tumor metabolism and nutritional status in pediatric oncology patients: two case
1016/j.clnu.2019.07.030. reports. J. Am. Coll. Nutr. 14 (2), 202–208.
Klement, R.J., Sweeney, R.A., 2016. Impact of a ketogenic diet intervention during Nencioni, A., Caffa, I., Cortellino, S., Longo, V.D., 2018. Fasting and cancer: molecular
radiotherapy on body composition: I. Initial clinical experience with six prospectively mechanisms and clinical application. Nat. Rev. Cancer 18 (11), 707–719.
studied patients. BMC Res. Notes 9, 143. Newman, J.C., Verdin, E., 2017. Beta-hydroxybutyrate: a signaling metabolite. Annu.
Korber, J., Pricelius, S., Heidrich, M., Muller, M.J., 1999. Increased lipid utilization in Rev. Nutr. 37, 51–76.
weight losing and weight stable cancer patients with normal body weight. Eur. J. Nguyen, X.C., Lee, W.W., Chung, J.H., Park, S.Y., Sung, S.W., Kim, Y.K., et al., 2007. FDG
Clin. Nutr. 53 (9), 740–745. uptake, glucose transporter type 1, and Ki-67 expressions in non-small-cell lung
Kubicek, G.J., Champ, C., Fogh, S., Wang, F., Reddy, E., Intenzo, C., et al., 2010. FDG-PET cancer: correlations and prognostic values. Eur. J. Radiol. 62 (2), 214–219.
staging and importance of lymph node SUV in head and neck cancer. Head Neck Nikoobakht, M., Shamshiripour, P., Azimi Nekoo, Z., Fallah Haghmohammadi, S., 2019.
Oncol. 2, 19. Elevated lactate and total protein levels in stereotactic brain biopsy specimen;

10
F. Bozzetti and Z. Stanga Critical Reviews in Oncology / Hematology 153 (2020) 103061

potential biomarkers of malignancy and poor prognosis. Arch. Iran. Med. 22 (3), 2013. Suppression of oxidative stress by beta-hydroxybutyrate, an endogenous his-
125–131. tone deacetylase inhibitor. Science (New York, NY). 339 (6116), 211–214.
Nolop, K.B., Rhodes, C.G., Brudin, L.H., Beaney, R.P., Krausz, T., Jones, T., et al., 1987. Shingler, E., Perry, R., Mitchell, A., England, C., Perks, C., Herbert, G., et al., 2019.
Glucose utilization in vivo by human pulmonary neoplasms. Cancer 60 (11), Dietary restriction during the treatment of cancer: results of a systematic scoping
2682–2689. review. BMC Cancer 19 (1), 811.
Norton, J.A., Burt, M.E., Brennan, M.F., 1980. In vivo utilization of substrate by human Sinclair, D.A., 2005. Toward a unified theory of caloric restriction and longevity reg-
sarcoma-bearing limbs. Cancer 45 (12), 2934–2939. ulation. Mech. Ageing Dev. 126 (9), 987–1002.
Osthus, R.C., Shim, H., Kim, S., Li, Q., Reddy, R., Mukherjee, M., et al., 2000. Skinner, R., Trujillo, A., Ma, X., Beierle, E.A., 2009. Ketone bodies inhibit the viability of
Deregulation of glucose transporter 1 and glycolytic gene expression by c-Myc. J. human neuroblastoma cells. J. Pediatr. Surg. 44 (1), 212–216 discussion 6.
Biol. Chem. 275 (29), 21797–21800. Solheim, T.S., Vagnildhaug, O.M., Laird, B.J., Balstad, T.R., 2019. Combining optimal
Ota, D.M., Nishioka, K., Grossie, B., Dixon, D., 1986. Erythrocyte polyamine levels during nutrition and exercise in a multimodal approach for patients with active cancer and
intravenous feeding of patients with colorectal carcinoma. Eur. J. Cancer Clin. Oncol. risk for losing weight: rationale and practical approach. Nutrition (Burbank, Los
22 (7), 837–842. Angeles County, Calif). 67–68 110541.
Pacelli, F., Bossola, M., Teodori, L., Trinca, M.L., Tortorelli, A., Rosa, F., et al., 2007. Stafford, P., Abdelwahab, M.G., Kim, D.Y., Preul, M.C., Rho, J.M., Scheck, A.C., 2010. The
Parenteral nutrition does not stimulate tumor proliferation in malnourished gastric ketogenic diet reverses gene expression patterns and reduces reactive oxygen species
cancer patients. South Afr. J. Clin. Nutr. 31 (6), 451–455. levels when used as an adjuvant therapy for glioma. Nutr Metab. 7, 74.
Park, S.I., Suh, D.S., Kim, S.J., Choi, K.U., Yoon, M.S., 2013. Correlation between biolo- Sun, W., Liu, Y., Glazer, C.A., Shao, C., Bhan, S., Demokan, S., et al., 2010. TKTL1 is
gical marker expression and F-fluorodeoxyglucose uptake in cervical cancer mea- activated by promoter hypomethylation and contributes to head and neck squamous
sured by positron emission tomography. Onkologie 36 (4), 169–174. cell carcinoma carcinogenesis through increased aerobic glycolysis and HIF1alpha
Patel, S., Alvarez-Guaita, A., Melvin, A., Rimmington, D., Dattilo, A., Miedzybrodzka, stabilization. Clin. Cancer Res. 16 (3), 857–866.
E.L., et al., 2019. GDF15 provides an endocrine signal of nutritional stress in mice and Takayama, K., Katakami, N., Yokoyama, T., Atagi, S., Yoshimori, K., Kagamu, H., et al.,
humans. Cell Metab. 29 (3), 707–718 e8. 2016. Anamorelin (ONO-7643) in Japanese patients with non-small cell lung cancer
Pfeiffer, T., Morley, A., 2014. An evolutionary perspective on the Crabtree effect. Front. and cachexia: results of a randomized phase 2 trial. Support. Care Cancer 24 (8),
Mol. Biosci. 1, 17. 3495–3505.
Phoenix, K.N., Vumbaca, F., Fox, M.M., Evans, R., Claffey, K.P., 2010. Dietary energy Tan-Shalaby, J.L., Carrick, J., Edinger, K., Genovese, D., Liman, A.D., Passero, V.A., et al.,
availability affects primary and metastatic breast cancer and metformin efficacy. 2016. Modified Atkins diet in advanced malignancies - final results of a safety and
Breast Cancer Res. Treat. 123 (2), 333–344. feasibility trial within the Veterans Affairs Pittsburgh Healthcare System. Nutr Metab.
Raffaghello, L., Lee, C., Safdie, F.M., Wei, M., Madia, F., Bianchi, G., et al., 2008. 13, 52.
Starvation-dependent differential stress resistance protects normal but not cancer Theodosakis, N., Held, M.A., Marzuka-Alcala, A., Meeth, K.M., Micevic, G., Long, G.V.,
cells against high-dose chemotherapy. Proc. Natl. Acad. Sci. U. S. A. 105 (24), et al., 2015. BRAF inhibition decreases cellular glucose uptake in melanoma in as-
8215–8220. sociation with reduction in cell volume. Mol. Cancer Ther. 14 (7), 1680–1692.
Rajaram, S., Baylink, D.J., Mohan, S., 1997. Insulin-like growth factor-binding proteins in van der Hulst, R.R., von Meyenfeldt, M.F., Deutz, N.E., Soeters, P.B., 1997. Glutamine
serum and other biological fluids: regulation and functions. Endocr. Rev. 18 (6), extraction by the gut is reduced in depleted [corrected] patients with gastrointestinal
801–831. cancer. Ann. Surg. 225 (1), 112–121.
Rice, Ml, van Rij, Am., 1987. Parenteral nutrition and tumour growth in the patient with van Solingen, C., Scacalossi, K.R., Moore, K.J., 2018. Long noncoding RNAs in lipid
complicated abdominal cancer. Aust. N. Z. J. Surg. 57 (6), 375–379. metabolism. Curr. Opin. Lipidol. 29 (3), 224–232.
Rieger, J., Bahr, O., Maurer, G.D., Hattingen, E., Franz, K., Brucker, D., et al., 2014. VanItallie, T.B., Nufert, T.H., 2003. Ketones: metabolism’s ugly duckling. Nutr. Rev. 61
ERGO: a pilot study of ketogenic diet in recurrent glioblastoma. Int. J. Oncol. 44 (6), (10), 327–341.
1843–1852. Veech, R.L., 2004. The therapeutic implications of ketone bodies: the effects of ketone
Rossi-Fanelli, F., Franchi, F., Mulieri, M., Cangiano, C., Cascino, A., Ceci, F., et al., 1991. bodies in pathological conditions: ketosis, ketogenic diet, redox states, insulin re-
Effect of energy substrate manipulation on tumour cell proliferation in parenterally sistance, and mitochondrial metabolism. Prostaglandins Leukot. Essent. Fatty Acids
fed cancer patients. Clin. Nutr. (Edinburgh, Scotland) 10 (4), 228–232. 70 (3), 309–319.
Rous, P., 1914. The influence of diet on transplanted and spontaneous mouse tumors. J. Veech, R.L., 2006. The determination of the redox states and phosphorylation potential in
Exp. Med. 20 (5), 433–451. living tissues and their relationship to metabolic control of disease phenotypes.
Saigusa, S., Toiyama, Y., Tanaka, K., Okugawa, Y., Fujikawa, H., Matsushita, K., et al., Biochem. Mol. Biol. Educ. 34 (3), 168–179.
2012. Prognostic significance of glucose transporter-1 (GLUT1) gene expression in Veech, R.L., Chance, B., Kashiwaya, Y., Lardy, H.A., Cahill Jr, G.F., 2001. Ketone bodies,
rectal cancer after preoperative chemoradiotherapy. Surg. Today 42 (5), 460–469. potential therapeutic uses. IUBMB Life 51 (4), 241–247.
San-Millan, I., Brooks, G.A., 2017. Reexamining cancer metabolism: lactate production Vesselle, H., Salskov, A., Turcotte, E., Wiens, L., Schmidt, R., Jordan, C.D., et al., 2008.
for carcinogenesis could be the purpose and explanation of the Warburg Effect. Relationship between non-small cell lung cancer FDG uptake at PET, tumor histology,
Carcinogenesis 38 (2), 119–133. and Ki-67 proliferation index. J. Thorac. Oncol. 3 (9), 971–978.
Scheck, A.C., Abdelwahab, M., Stafford, P., Kim, D.-Y., Iwai, S., Preul, M.C., et al., 2010. Voss, M., Marlies, Wagner., von Mettenheim, N., et al. ERGO2: A prospective randomized
Mechanistic studies of the ketogenic diet as an adjuvant therapy for malignant trial of calorie restricted ketogenic diet and fasting in addition to re-irradiation for
gliomas. AACR. malignant glioma [published online ahead of print, 2020 Jun 30]. Int J Radiat Oncol
Schmidt, M., Pfetzer, N., Schwab, M., Strauss, I., Kammerer, U., 2011. Effects of a keto- Biol Phys. 2020;S0360-3016(20)31308-0. doi:10.1016/j.ijrobp.2020.06.021.
genic diet on the quality of life in 16 patients with advanced cancer: a pilot trial. Walenta, S., Salameh, A., Lyng, H., Evensen, J.F., Mitze, M., Rofstad, E.K., et al., 1997.
Nutr. Metab. 8 (1), 54. Correlation of high lactate levels in head and neck tumors with incidence of metas-
Schroeder, U., Himpe, B., Pries, R., Vonthein, R., Nitsch, S., Wollenberg, B., 2013. Decline tasis. Am. J. Pathol. 150 (2), 409–415.
of lactate in tumor tissue after ketogenic diet: in vivo microdialysis study in patients Walenta, S., Wetterling, M., Lehrke, M., Schwickert, G., Sundfor, K., Rofstad, Ek, et al.,
with head and neck cancer. Nutr. Cancer 65 (6), 843–849. 2000. High lactate levels predict likelihood of metastases, tumor recurrence, and
Schuurbiers, O.C., Meijer, T.W., Kaanders, J.H., Looijen-Salamon, M.G., de Geus-Oei, L.F., restricted patient survival in human cervical cancers. Cancer Res. 60 (4), 916–921.
van der Drift, M.A., et al., 2014. Glucose metabolism in NSCLC is histology-specific Walenta, S., Chau, T.V., Schroeder, T., Lehr, H.A., Kunz-Schughart, L.A., Fuerst, A., et al.,
and diverges the prognostic potential of 18FDG-PET for adenocarcinoma and squa- 2003. Metabolic classification of human rectal adenocarcinomas: a novel guideline
mous cell carcinoma. J. Thorac. Oncol. 9 (10), 1485–1493. for clinical oncologists? J. Cancer Res. Clin. Oncol. 129 (6), 321–326.
Schwartz, G.F., Green, H.L., Bendon, M.L., Graham 3rd, W.P., Blakemore, W.S., 1971. Wang, L., Xiong, H., Wu, F., Zhang, Y., Wang, J., Zhao, L., et al., 2014. Hexokinase 2-
Combined parenteral hyperalimentation and chemotherapy in the treatment of dis- mediated Warburg effect is required for PTEN- and p53-deficiency-driven prostate
seminated solid tumors. Am. J. Surg. 121 (2), 169–173. cancer growth. Cell Rep. 8 (5), 1461–1474.
Schwartz, K., Chang, H.T., Nikolai, M., Pernicone, J., Rhee, S., Olson, K., et al., 2015. Wang, Y., Zhang, X., Wang, Z., Hu, Q., Wu, J., Li, Y., et al., 2018. LncRNA-p23154 pro-
Treatment of glioma patients with ketogenic diets: report of two cases treated with an motes the invasion-metastasis potential of oral squamous cell carcinoma by reg-
IRB-approved energy-restricted ketogenic diet protocol and review of the literature. ulating Glut1-mediated glycolysis. Cancer Lett. 434, 172–183.
Cancer Metab. 3, 3. Warburg, O., Wind, F., Negelein, E., 1927. The metabolism of tumors in the body. J. Gen.
Selberg, O., McMillan, D.C., Preston, T., Carse, H., Shenkin, A., Burns, H.J., 1990. Physiol. 8 (6), 519–530.
Palmitate turnover and its response to glucose infusion in weight-losing cancer pa- Ward, P.S., Thompson, C.B., 2012. Metabolic reprogramming: a cancer hallmark even
tients. Clin. Nutr. (Edinburgh, Scotland) 9 (3), 150–156. warburg did not anticipate. Cancer Cell 21 (3), 297–308.
Seyfried, T.N., Shelton, L., Arismendi-Morillo, G., Kalamian, M., Elsakka, A., Maroon, J., Wei, S., Fan, Q., Yang, L., Zhang, X., Ma, Y., Zong, Z., et al., 2017. Promotion of glycolysis
et al., 2019. Provocative question: should ketogenic metabolic therapy become the by HOTAIR through GLUT1 upregulation via mTOR signaling. Oncol. Rep. 38 (3),
standard of care for glioblastoma? Neurochem. Res. 44 (10), 2392–2404. 1902–1908.
Shaw, J.H., Wolfe, R.R., 1987. Fatty acid and glycerol kinetics in septic patients and in Weinberg, F., Chandel, N.S., 2009. Reactive oxygen species-dependent signaling regulates
patients with gastrointestinal cancer. The response to glucose infusion and parenteral cancer. Cell. Mol. Life Sci. 66 (23), 3663–3673.
feeding. Ann. Surg. 205 (4), 368–376. Westin, T., Stein, H., Niedobitek, G., Lundholm, K., Edstrom, S., 1991. Tumor cytokinetic
Shelton, L.M., Huysentruyt, L.C., Mukherjee, P., Seyfried, T.N., 2010. Calorie restriction response to total parenteral nutrition in patients with head and neck cancers. Am. J.
as an anti-invasive therapy for malignant brain cancer in the VM mouse. ASN Neuro 2 Clin. Nutr. 53 (3), 764–768.
(3), e00038. Wieman, H.L., Wofford, J.A., Rathmell, J.C., 2007. Cytokine stimulation promotes glu-
Shim, H., Dolde, C., Lewis, B.C., Wu, C.S., Dang, G., Jungmann, R.A., et al., 1997. c-Myc cose uptake via phosphatidylinositol-3 kinase/Akt regulation of Glut1 activity and
transactivation of LDH-A: implications for tumor metabolism and growth. Proc. Natl. trafficking. Mol. Biol. Cell 18 (4), 1437–1446.
Acad. Sci. U. S. A. 94 (13), 6658–6663. Winter, S.F., Loebel, F., Dietrich, J., 2017. Role of ketogenic metabolic therapy in ma-
Shimazu, T., Hirschey, M.D., Newman, J., He, W., Shirakawa, K., Le Moan, N., et al., lignant glioma: a systematic review. Crit. Rev. Oncol. Hematol. 112, 41–58.

11
F. Bozzetti and Z. Stanga Critical Reviews in Oncology / Hematology 153 (2020) 103061

Wise, D.R., DeBerardinis, R.J., Mancuso, A., Sayed, N., Zhang, X.Y., Pfeiffer, H.K., et al., Glucose deprivation contributes to the development of KRAS pathway mutations in
2008. Myc regulates a transcriptional program that stimulates mitochondrial gluta- tumor cells. Science (New York, NY) 325 (5947), 1555–1559.
minolysis and leads to glutamine addiction. Proc. Natl. Acad. Sci. U. S. A. 105 (48), Zeng, Y., Ren, K., Zhu, X., Zheng, Z., Yi, G.Long, 2018. Noncoding RNAs: advances in lipid
18782–18787. metabolism. Adv. Clin. Chem. 87, 1–36.
Wofford, J.A., Wieman, H.L., Jacobs, S.R., Zhao, Y., Rathmell, J.C., 2008. IL-7 promotes Zhao, Y., Liu, Y., Lin, L., Huang, Q., He, W., Zhang, S., et al., 2018. The lncRNA MACC1-
Glut1 trafficking and glucose uptake via STAT5-mediated activation of Akt to support AS1 promotes gastric cancer cell metabolic plasticity via AMPK/Lin28 mediated
T-cell survival. Blood. 111 (4), 2101–2111. mRNA stability of MACC1. Mol. Cancer 17 (1), 69.
Wong, N., Ojo, D., Yan, J., Tang, D., 2015. PKM2 contributes to cancer metabolism. Zheng, M., Cao, M.X., Yu XH, Li L., Wang, K., Wang, S.S., et al., 2019. STAT3 promotes
Cancer Lett. 356 (2 Pt A), 184–191. invasion and aerobic glycolysis of human oral squamous cell carcinoma via inhibiting
Yang, C., Sudderth, J., Dang, T., Bachoo, R.M., McDonald, J.G., DeBerardinis, R.J., 2009. FoxO1. Front. Oncol. 9, 1175.
Glioblastoma cells require glutamate dehydrogenase to survive impairments of glu- Zhou, W., Mukherjee, P., Kiebish, M.A., Markis, W.T., Mantis, J.G., Seyfried, T.N., 2007.
cose metabolism or Akt signaling. Cancer Res. 69 (20), 7986–7993. The calorically restricted ketogenic diet, an effective alternative therapy for malig-
Yang, X., Yao, B., Niu, Y., Chen, T., Mo, H., Wang, L., et al., 2019. Hypoxia-induced nant brain cancer. Nutr. Metab. 4, 5.
lncRNA EIF3J-AS1 accelerates hepatocellular carcinoma progression via targeting Zuccoli, G., Marcello, N., Pisanello, A., Servadei, F., Vaccaro, S., Mukherjee, P., et al.,
miR-122-5p/CTNND2 axis. Biochem. Biophys. Res. Commun. 518 (2), 239–245. 2010. Metabolic management of glioblastoma multiforme using standard therapy
Yun, J., Rago, C., Cheong, I., Pagliarini, R., Angenendt, P., Rajagopalan, H., et al., 2009. together with a restricted ketogenic diet: case report. Nutr Metab. 7, 33.

12

You might also like