You are on page 1of 15

REVIEWS

Developing dietary interventions


as therapy for cancer
Samuel R. Taylor   1,2,3, John N. Falcone1, Lewis C. Cantley   2
and Marcus D. Goncalves   1,2 ✉
Abstract | Cancer cells acquire distinct metabolic preferences based on their tissue of origin,
genetic alterations and degree of interaction with systemic hormones and metabolites.
These adaptations support the increased nutrient demand required for increased growth and
proliferation. Diet is the major source of nutrients for tumours, yet dietary interventions lack
robust evidence and are rarely prescribed by clinicians for the treatment of cancer. Well-controlled
diet studies in patients with cancer are rare, and existing studies have been limited by nonspecific
enrolment criteria that inappropriately grouped together subjects with disparate tumour and
host metabolic profiles. This imprecision may have masked the efficacy of the intervention for
appropriate candidates. Here, we review the metabolic alterations and key vulnerabilities that
occur across multiple types of cancer. We describe how these vulnerabilities could potentially be
targeted using dietary therapies including energy or macronutrient restriction and intermittent
fasting regimens. We also discuss recent trials that highlight how dietary strategies may be
combined with pharmacological therapies to treat some cancers, potentially ushering a path
towards precision nutrition for cancer.

Diet plays a role in virtually all aspects of cancer includ- studies in mice, such as those by Hopkins et al., Caffa et al.
ing tumour initiation, progression and response to treat- and Maddocks et al., have shown that dietary interven-
ment. For example, estimates suggest that up to one-​third tions enhance anticancer therapy and improve cancer
of the most common cancers are preventable, in part, outcomes12–14. The translation of these findings is well
through dietary modification1–3. These associations were underway with early-​phase clinical data supporting their
established using large-​scale population-​based observa- feasibility and safety15,16. Over the next 5 years, random­
tional cohort studies, which provide valuable information ized controlled trials will be conducted to establish the
about correlations but cannot assess causation and suf- importance of diet on clinical outcomes in patients with
fer from the notorious difficulty of accurately recording cancer. In this Review, we describe the key genetic and
dietary intake4,5. Controlled, mechanistic studies in pre- metabolic features of tumours and their host tissues that
clinical models have greatly improved our understanding should make them vulnerable to dietary interventions.
of how tissues utilize nutrients and the impact diet may We assess the state of the field and offer suggestions on
play in tumour initiation, progression and treatment how to make precision nutrition a reality for cancer
response. These studies have shown that diet can supply treatment.
the tumour with nutrients such as sugars and lipids that
1
Division of Endocrinology,
Department of Medicine,
can be used for tumour growth6; modulate systemic hor- Metabolic vulnerabilities of cancer
Weill Cornell Medicine, mones such as leptin, insulin, insulin-​like growth factor 1 General pathways of nutrient metabolism
New York, NY, USA. (IGF1) and oestradiol that promote tumour cell pro- The reprogramming of cell metabolism is a hallmark of
2
Meyer Cancer Center, Weill liferation and survival7,8; and support the development cancer17. Transformed cells adapt to a state of unregu-
Cornell Medicine, New York, of obesity, which creates a milieu that favours tumour lated cell growth and proliferation by upregulating nutri-
NY, USA.
initiation and progression8. As the links between dietary ent uptake and anabolic processes (Fig. 1). Sugars, amino
3
Weill Cornell/Rockefeller/ factors and tumour initiation have been well reviewed acids and fats are the basic dietary nutrients that support
Sloan Kettering Tri-​I MD–PhD
program, New York, NY, USA.
and summarized elsewhere9–11, our focus in this Review the growth of cancer cells.
✉e-​mail: mdg9010@ is the impact of diet on tumour progression and how this Glucose, the most extensively studied nutrient in
med.cornell.edu information is being translated into anticancer therapy. cancer biology, is a primary energy source for cells and
https://doi.org/10.1038/ We are on the precipice of a revolution in the way we is obtained from the digestion of dietary carbohydrates
s41568-022-00485-​y treat patients with cancer. Several rigorous preclinical or generated de novo by anabolic processes in the liver

NATuRE REVIEwS | Cancer

0123456789();:
Reviews

Electromotive force
or kidney. Once imported via glucose transporters, efficient way to generate ATP per unit of glucose as com-
The electric potential glucose can be metabolized in cells in two main ways. The pared with glycolysis, but less efficient than glycolysis
generated by the position first and ancestrally older path is glycolysis. Glycolysis when considered per unit of oxygen18.
of charged molecules, such takes place in the cytosol and breaks down glucose to yield The vast majority of ATP comes from the coupling of
as when partitioned across
a membrane.
ATP, reducing equivalents, such as NADH, and pyruvate. glycolysis with oxidative phosphorylation but this link is
The second, oxidative phosphorylation, uses energy perturbed in two scenarios. The first is hypoxia, where
ATP synthase from the oxidation of reducing equivalents to create an oxygen becomes limiting and the final electron transfer
A mitochondrial enzyme electromotive force across the mitochondrial inner mem- of oxidative phosphorylation is halted. Hypoxic cells
that phosphorylates
brane, reducing and consuming oxygen in the process. upregulate glycolysis to generate ATP and reduce the
ADP to make ATP.
This potential difference dissipates through ATP synthase overflow of NADH18. The second scenario occurs in cer-
to generate ATP. Oxidative phosphorylation is a more tain rapidly proliferating cells such as T cells, embryonic
Fructose Glucose Lipoproteins
GLUT5 GLUT1–GLUT4 LDLR

• ROS mitigation
KHK • Nucleotide synthesis
SDH HK
F1P Glycogen G6P PPP Cholesterol
SLC1A4 Glycine G6PD
Lipid droplet
ALDO Fatty acid
Glycine reservoir
and serine Lipid membrane
DHAP Glycerol-3-P triacylglycerides
Glyceraldehyde Glyceraldehyde-3-P Oleate
SCD
Serine
PHGDH O2
3-Phosphoglycerate FAO Stearate Fatty
Amino acids acids
and nucleotides αKG Glutamate CPT1 CD36
ATP ATP Palmitate
PEP
FASN
PKM2
LDH
Lactate Lactate Pyruvate Malonyl-CoA
PDH ACC
MCTs Aspartate
ACLY
Acetyl-CoA Acetyl-CoA Acetate
Proteins and Oxaloacetate
nucleotides ACSS1 ACSS2
Citrate Citrate MCTs
Steroid hormone
CO2 IDH2 IDH1 synthesis
αKG
ATP
O2 Epigenetic dysregulation
mtIDH1/2
αKG (D)-2HG
ROS
BCATs
Propionyl-CoA
GSH Glutamate
Proteins and LATs
GSS
Cysteine nucleotides
Glycine GLS

Glutamine Arginine

BCAAs,
SLC7A11 SLC1A5 ATs methionine
Cystine Glutamate Glutamine Arginine and threonine

Fig. 1 | nutrient utilization in cancer. Various pathways mediate oxidation; FASN, fatty acid synthase; G6PD, glucose-6-phosphate
conversion of extracellular sugars, fats and amino acids into energy or dehydrogenase; GLS, glutaminase; GLUT, glucose transporter; GSS,
building blocks for anabolic processes. Enzymes critical to and/or frequently glutathione synthetase; (mt)IDH, (mutated) isocitrate dehydrogenase; KHK,
upregulated in cancer in these pathways are shown, colour coded ketohexokinase; LATs, L-type amino acid transporters; LDH, lactate
according to their primary nutrient association: fructose (beige), glucose dehydrogenase; LDLR, low-density lipoprotein receptor; MCTs,
(pink), lipid uptake and degradation (yellow) and amino acid metabolism mono-carboxylase transporters; PDH, pyruvate dehydrogenase; PHGDH,
(blue). ACC, acetyl-​CoA carboxylase; ACLY, ATP citrate lyase; ACSS, phosphoglycerate dehydrogenase; PKM2, pyruvate kinase isoform M2; PPP,
acetyl-CoA synthetase; ALDO, aldolase; ATs, arginine transporters; pentose-phosphate pathway; SCD, stearoyl-CoA desaturase; SDH, sorbitol
BCAAs, branched-chain amino acids; BCATs, branched-chain amino dehydrogenase; SLC1A4, solute carrier family 1 member 4; SLC1A5, solute
acid transaminases; CPT1, carnitine palmitoyl transferase I; FAO, fatty acid carrier family 1 member 5; SLC7A11, solute carrier family 7 member 11.

www.nature.com/nrc

0123456789();:
Reviews

Brain Lung
Limited serine, glycine Variable O2 exposure
1
High uptake of glucose, High uptake of lactate, fatty acids
lactate, glutamine

Breast Pancreas
2 High synthetic burden
Local fat reservoir
3
High uptake of fatty acids, High uptake of amino acids
lipoproteins, methionine (glutamine, alanine, BCAAs)
4
Sensitive to insulin/IGF1, 5 Intestine
steroid hormones 6
• Steep O2 gradients
• Direct exposure to dietary
Liver 7 nutrients and microbiota-
Exposed to intestinal/pancreatic 8 derived metabolites
metabolites via portal circulation
High uptake of sugars, glutamine,
High uptake of lactate, organic short-chain fatty acids
acids, bile salts, short-chain
fatty acids Prostate
Local source of fructose and citric
Sensitive to insulin/IGF1, acid in seminal fluid
steroid hormones
High uptake of fatty acids,
Uterus/endometrium citrate, lactate
Sensitive to insulin/IGF1,
steroid hormones Sensitive to steroid hormones

Local tissue environment/function factors Nutrient uptake preference Hormone sensitivity

Fig. 2 | Tissue-specific metabolism. Notable metabolic features of various tissues. Anatomic location of these tissues
as well as metabolic demands of their normal function may impart vulnerabilities that can be targeted with diet and/or
pharmacotherapy. See main text for relevant references. BCAAs, branched-​chain amino acids; IGF1, insulin-​like
growth factor 1.

stem cells and tumour cells19–21. Here, regardless of oxy- off for various anabolic processes such as the synthesis of
gen levels, cells typically exhibit high rates of glycolysis fatty acids35. In contrast to glutamine, the branched-​chain
and lactic fermentation. Lactate was long thought to be a amino acids (BCAAs) (leucine, isoleucine and valine) are
mere waste product of glycolysis, but is now recognized essential amino acids primarily obtained from dietary
as an important fuel source for human tumours and sources. Studies in mice show that these can be taken
healthy tissues22–24. In fact, stable isotope tracing studies up by many tissues and tumours to be incorporated into
in fasted mice reveal that lactate incorporation into newly synthesized proteins or oxidized for fuel36,37.
tricarboxylic acid (TCA) cycle intermediates surpasses Fatty acids are a dense source of energy that can be
that of glucose in all tissues except the brain24,25. obtained from the diet and stored in the body as triglyce­
In addition to glucose and lactate, fructose also plays rides. In times of low food intake, mammals catabolize
an important role in the metabolism of many different triglycerides and release the resulting fatty acids and
tissues. Although the small intestine and liver see the glycerol. Glycerol can be used by cells as a glycolytic,
largest concentrations of dietary fructose, serum fruc- gluconeogenic or lipogenic substrate. Fatty acids can
tose rises significantly after fructose consumption and is be oxidized to generate ATP, reused in the synthesis of
metabolized by tissues such as the kidney, prostate and other lipid moieties such as phospholipids and choles-
bone marrow26–29. Inside the cell, fructose can be cleaved terol esters or stored in intracellular lipid droplets. When
into glycolytic intermediates and oxidized for energy oxidized by the liver, fatty acids generate ketone bodies,
production. Despite using many similar pathways to which are released into circulation and fed into the TCA
glucose, fructose metabolism is uniquely regulated cycle in certain tissues25. These processes are frequently
and often results in different downstream end points30. upregulated in rapidly proliferating tissues such as
Studies in mice suggest that fructose can reprogramme tumours38–40.
metabolic pathways for anabolic metabolism and cell
survival31, a role that may be particularly important in Tissue-​specific metabolism
Ketone bodies solid tumours32–34. Cancer cells acquire distinct metabolic preferences based
Metabolites such as Other nutrients such as amino acids serve as build- on their tissue of origin, genetic alterations and degree
β-​hydroxybutyrate and ing blocks for anabolic processes. Glutamine is a of interaction with systemic hormones and metabolites.
acetoacetate, which are non-essential amino acid arising from dietary sources, The tissue of origin has a particularly strong effect on
produced during hepatic
fatty acid oxidation and
muscle degradation or de novo synthesis pathways. Via its metabolite abundance and metabolic gene expression in
can be used as energy conversion to α-​ketoglutarate, glutamine provides carbon tumours owing to engrained, lineage-​dependent prefer-
substrates by some tissues. for TCA cycle intermediates that can then be siphoned ences for sugars, fats and amino acids41–44 (Fig. 2). For the

NATuRE REVIEwS | Cancer

0123456789();:
Reviews

sake of brevity, this Review focuses on the metabolism Breast. Mammary glands within breast tissue have a
of a subset of tissues that have a high predilection for high capacity to synthesize large amounts of macro­
malignant transformation as well as strong literature molecules, which get exported into milk. The substrates
supporting a relationship with dietary nutrients. For the for this process are diet-​derived and include glucose,
interested reader, excellent reviews are available summa- fatty acids, lipoproteins and amino acids (especially
rizing what is known about how diet and tumour metab- methionine)64. Much nutrient uptake by the breast is
olism relate in other cancer types, for example gastric regulated by insulin, IGF1 and steroid hormones such as
and kidney cancers45,46. oestrogens and progesterone. For example, fluorodeoxy-
glucose (FDG) uptake in human breast tissue increases
Brain. The brain is a unique organ that depends on the with progesterone levels, and uptake and production of
uptake and oxidation of glucose and/or ketone bodies lipids and proteins are strongly regulated by insulin65–68.
and is also separated from systemic circulation by the Dysregulation of the pathways involved in hormone
selectively permeable blood–brain barrier. High glu- sensing and nutrient uptake in the breast can promote
cose utilization is driven, in part, by expression of the cancer development. For example, the PI3K pathway is
gene encoding the transporter solute carrier family 2 typically hyperactivated in human breast tumours via
member 3 (SLC2A3), which has a high affinity and mutations in the gene for the main catalytic subunit,
trans­port capacity for glucose47. Interestingly, mouse PIK3CA (ref.69). PI3K is a lipid kinase that responds
studies reveal that there is marked heterogeneity in to environmental growth signals such as insulin and
glucose uptake within different regions of the brain48. IGF1 to promote proliferation and nutrient uptake70.
Furthermore, a large body of evidence including tracer Studies with patient-​derived cell lines show that hyper-
imaging studies in mice and humans indicates that neu- activation of PI3K is one way that oestrogen receptor
rons may prefer to use lactate generated by local astro- (ER)-​positive tumours may evade hormonal therapy71.
cytes as a primary fuel49. Mouse and rat studies suggest Correspondingly, data from mouse models and humans
that the brain is also capable of metabolizing ketone treated with PI3K inhibitors show that tumours upregu-
bodies during fasting, although the uptake and usage is late ER signalling as an escape strategy72. These findings
also spatially heterogeneous50–52. Fatty acids, meanwhile, have prompted clinicians to begin targeting both PI3K
are seldom used by neuronal cells to meet energetic and ER — an approach that is proving effective in some
demands25,53. Similarly, amino acids such as glutamine patients with breast cancer73.
are rarely oxidized by brain cells25. Glutamate is an exci­ The molecular features of breast tumours impact
tatory neurotransmitter than can cause neurotoxicity if nutrient uptake and metabolism. Human imaging studies
it accumulates, so it is rapidly converted to glutamine corroborate preclinical models showing that ER-​positive
by astrocytes and then transported back to neurons49. tumours are relatively oxidative and consume lactate
Primary brain tumours adapt metabolically to grow and citrate, whereas triple-​negative breast tumours are
and survive. For example, many brain tumours express more glycolytic lactate exporters74. The expression of
higher levels or different isoforms of glycolytic enzymes oestrogen-related receptors (ERRs), orphan nuclear
(for example, hexokinase, pyruvate kinase, pyruvate receptors with structural similarity to ER, can also change
dehydrogenase (PDH)), which provide them with specific breast tumour metabolism. For example, human
advantages in the brain’s unique environment (reviewed genomic studies and in vitro models indicate that ERRα
in detail elsewhere49). Some of these unique features activity promotes the expression of myriad metabolic
may be exploited for therapy or diagnosis. For example, enzymes and confers an aggressive glycolytic phenotype
in vitro studies indicate that depriving brain tumours of with a worse prognosis75. Human genomic studies and
glucose may be a useful therapeutic strategy as normal xenograft mouse models also show that triple-​negative
neurons, but not glioma cells, are capable of surviving breast tumours depend on de novo synthesis of serine,
on ketone bodies54. A subset of particularly aggressive which is limiting in the tumour microenvironment and
brain tumours promote glycolysis by downregulating can be further depleted via diet76–78.
the mitochondrial pyruvate transporter subunit MPC1, Fatty acid metabolism also appears important for
which makes cells dependent upon glutamine uptake the development of certain molecular subtypes of breast
in vitro49,55,56. Mutations in isocitrate dehydrogenase cancer. For example, compared against breast tissue
(IDH1 or IDH2) occur in 70% of intermediate-​grade glio- from healthy women, normal tissue from women who
mas and affect glioma sensitivity to nutrient deprivation57; later developed cancer showed upregulated fatty acid
in  vitro tracing studies suggest that these tumours uptake and transport, lipolysis, lipid peroxidation and
particularly depend on glutamine58–60 (Fig. 1). epithelium–adipose tissue crosstalk79. Uptake of exog-
Nutrient availability also appears to affect the growth enous fatty acids portends a worse prognosis and is
of metastatic tumours within the brain. For example, the typical of aggressive, triple-​negative tumours80. Fatty
brain has relatively low availability of serine and fatty acids acid metabolism may also be perturbed in local adipose
Triple-​negative breast
tumours relative to serum, and mouse models indicate that breast deposits in the breast. In vitro co-​culture studies show
Breast tumours with low levels cancer metastases to the brain have increased reliance on that adipocytes can deliver fatty acids to tumour cells and
of oestrogen receptor (ER), de novo generation of these components61,62. Furthermore, induce gene expression changes that enhance tumour
progesterone receptor and nuclear magnetic resonance (NMR) labelling studies of cell migration81,82. Adipocytes also generate oestrogen via
HER2 overexpression and/or
amplification. Typically,
resected human tumours reveal that metastases from a wide aromatase, which may explain why oestrogen levels are
these tumours carry a worse variety of primary sites can acquire the ability to oxidize much higher in benign human breast tissue and tumours
prognosis than other types. acetate as a fuel source when they arrive in the brain63. than in serum83.

www.nature.com/nrc

0123456789();:
Reviews

Proliferative phase of the


Endometrium. Similar to the breast, the endometrium important for prostate cancers, as suppressing their
endometrial cycle is a hormone-​responsive tissue with high fluctuations uptake slows tumour progression in mouse models95,96.
The phase of the endometrial in metabolic demand. Studies from mouse and human
cycle in which the endometrial stromal cells show that glucose trans- Lung. Stable isotope labelling studies in healthy pigs
endometrial cells rapidly
porter expression and glucose uptake are positively show that lung metabolism is notable for relatively
proliferate in preparation
for possible implantation regulated by progesterone and negatively regulated by high consumption of hydroxybutyrate and acetoacetate
of a fertilized embryo. oestrogen84. Moreover, insulin stimulates the growth of (ketone bodies), glutamine and saturated fatty acids97.
the endometrial mucosa, especially in the proliferative Nutrient uptake in the lung is not particularly sensitive
phase of the endometrial cycle where IGF1 signalling is to systemic hormones. For example, there are minimal
essential85. In mouse models, cell-​autonomous activa- changes in the contribution of glucose and lactate to
tion of the PI3K pathway is sufficient for the initiation TCA cycle intermediates with fasting and refeeding, as
of endometrial tumours86, and the PI3K pathway is fre- compared with other tissues25.
quently activated in human endometrial carcinoma6,87. The lung is an excellent example of a tissue where
More than 90% of human endometrial tumours have local effects likely alter tumour metabolic phenotypes.
genetic alterations in the PI3K signalling pathway, often As in many other tumours, mutations in primary lung
by activating mutations in PIK3CA, or loss-​of-​function tumours occur in pathways that are associated with
mutations in PTEN, the gene encoding a phosphatase increased glucose uptake and flux to anabolic pathways.
that directly opposes PI3K (refs6,87). These alterations These mutations include genes in the epidermal growth
likely underlie the clinical associations of endometrial factor receptor (EGFR)–PI3K pathway, oncogenes such
cancer with biomarkers of hyperinsulinemia such as as KRAS and MYC, and tumour suppressor genes such as
fasting glucose, insulin, c-​peptide (insulin production), STK11 (ref.98). However, lung tumours with these altera­
haemoglobin A1C (chronic hyperglycaemia) and dietary tions often have different metabolic phenotypes com-
glycaemic load6. pared with genetically similar tumours in other tissues.
This distinction may be due to the unique features of
Prostate. As with the breast and endometrium, the the lung environment, which offer a set of metabolic
prostate is highly sensitive to sex hormones. In partic­ advantages to primary and metastatic tumours alike.
ular, testosterone drives glucose uptake, which can be For example, studies in mouse models show that the
detected via FDG-​PET and reversed with oestrogen lung environment may better allow pyruvate and proline
administration in rats88. In vitro and mouse models also metabolism and, thereby, preferentially support breast
show that testosterone significantly accelerates incorpo- cancer metastasis99,100. Another example comes from
ration of amino acids into protein and of acetate into in-​human stable-​isotope tracing studies that found that
fatty acids and citric acid89. These changes help support non-​small-​cell lung cancer (NSCLC) tumours maintain
the primary biosynthetic role of the prostate to produce high levels of glucose oxidation and can use a wide variety
seminal fluid, which is rich in citric acid and hydrolytic of TCA substrates including, possibly, lactate101. In this
enzymes arising from specialized epithelial cells. These study, tumour metabolism was highly heterogeneous
cells are highly glycolytic and have diminished capacity between and within tumours, suggesting that variations
for oxidative phosphorylation to enable them to gene­ in the tumour environment within the same tissue also
rate and export citrate90. Seminal fluid is also rich in alter metabolism101. A final illustration of the tumour
fructose, and studies from human tissue suggest that site altering the metabolic phenotype comes from mouse
this fructose is generated de novo from glucose via the studies that show that KRAS-​mutant pancreatic tumours
polyol pathway91. This pathway uses aldose reductase to avidly scavenge their microenvironment for nutrients via
reduce glucose to sorbitol, and then sorbitol dehydro- macropinocytosis, but KRAS-​mutant NSCLC tumours
genase (SDH) that oxidizes sorbitol to fructose (Fig. 1). take up comparatively less — even when derived within
Interestingly, SDH is an androgen-​regulated gene that the same organism37,102.
is expressed in benign and malignant human prostate Some metabolic features of lung tumours may prove
cancers, in addition to the seminal vesicles92. to be metabolic vulnerabilities. For example, human
As with most other tumour types, cancerous trans- lung tumours express high levels of fructolytic enzymes
formation increases nutrient uptake and utilization relative to normal lung tissue, and inhibiting fructose
in prostate cancer. Interestingly, mouse and human uptake reduces tumour growth in xenograft models34.
tissue studies show that prostate cancer cells regain the Glutamine dependence has also been reported in some
ability to perform oxidative phosphorylation during preclinical KRAS-​driven tumour studies, with cells
transformation90. This adaptation allows these tumours carrying KRAS-​G12V being less dependent on glu-
to readily consume and oxidize locally produced nutri- tamine than cells expressing KRAS-​G12(C/D) (ref.103).
ents such as citrate and lactate74,90. Moreover, emerging However, other studies in mouse models suggest that
evidence suggests that fructose can be a key substrate glutamine metabolism is dispensable for fuelling the
for prostate cancer cells. For example, human primary TCA cycle when glucose is present104,105. KRAS-​mutated
prostate tumours have high expression of SLC2A5, tumours may also be sensitive to therapies targeting
the main fructose transporter28. Fructose may arise BCAA and fatty acid metabolism. BCAA uptake is
from dietary sources or the tumours may gain access elevated in NSCLC tumours versus normal lung, con-
to fructose-​dense fluid during seminal vesicle inva- sistent with low serum levels early in clinical disease,
sion, which confers a worse prognosis in humans even and deletion of the BCAA transporter impairs tumour
above other local invasion93,94. Fatty acids may also be formation in mice 37,106. Fatty acid metabolism may

NATuRE REVIEwS | Cancer

0123456789();:
Reviews

also be a promising target, and at least one enzyme, Traditionally, the intestine has been seen as mainly a
acyl-coenzyme A synthetase long-​chain family member 3 transporter of dietary nutrients, but recent work has
(ACSL3), is highly expressed in human NSCLC and is highlighted its more active role in metabolism. For
essential for KRAS-​mutant lung cancer tumorigenesis example, the mouse intestinal epithelium converts low
in mice107. doses of dietary fructose into glucose and organic acids
that are subsequently delivered to the portal blood
Pancreas. The primary role of the pancreas is anabolic: and liver27. In vivo labelling studies in mice and pigs
to synthesize and secrete hormones and enzymes. This also show that the intestines are the primary site for
process requires a large influx of amino acids, which production of the body’s short-​chain fatty acids such
serve as both building blocks for protein synthesis and as propionate, butyrate and acetate, which are mainly
fuel for TCA cycle-​dependent amino acid production. generated by gut microbiota97,116. In addition to pro-
Out of 11 organs thoroughly profiled in mice and pigs, ducing nutrients for the body, the intestines consume
the pancreas shows the greatest use of amino acids25,97. circulating nutrients such as glucose and glutamine.
These amino acids are consumed and repurposed by In fact, mouse and porcine labelling studies also show
zymogen-​producing acinar cells25,36. that the small intestine is one of the primary sites of
Despite its role as a major endocrine organ and expo- glucose disposal in the fed state and of ketone body
sure to high levels of insulin, the uptake of nutrients by oxidation while fasting25,97.
the pancreas is not particularly sensitive to systemic Colorectal cancer (CRC) is the most common cancer
hormones. In fact, insulin-​mediated glucose uptake arising from the intestine, and its metabolism reflects
contributes little to TCA intermediates in mouse pan- its driving mutations, nutrient availability and tissue of
creas. Instead, fatty acids and ketone bodies are used origin. Most CRCs arise from sporadic mutations in the
more readily, especially in times of fasting25. Despite WNT, PI3K and KRAS pathways13–15. WNT signalling
these findings, systemic insulin has been shown to pro- is hyperactive in 93% of CRC tumours and is associated
mote the formation of precancerous lesions in the mouse with impaired oxidative metabolism, increased pentose-
pancreas108. These data suggest that the role of insulin in phosphate pathway (PPP) flux and increased fatty acid
pancreas tumorigenesis may be independent of its ability synthesis in in vitro studies, mouse xenograft models
to stimulate glucose uptake. and human tumour samples117,118. PI3K pathway muta-
Mutations in KRAS are common in human pancreatic tions confer a dependence upon glutamine uptake
ductal adenocarcinomas (PDACs), and these tumours and metabolism, which have been effectively targeted
have several familiar and several unique metabolic to reduce tumour growth in mouse models119,120. KRAS
features that may prove to be targetable vulnerabilities. mutations confer enhanced glucose transporter expres-
For example, in vitro labelling and mouse tumour genetic sion and improved cell survival in low-​glucose con-
analyses indicate enhanced glucose uptake, flux through ditions in human CRC-​derived cell lines and mouse
glycolysis and diversion of carbon into the hexosamine xenograft tumours121. Emerging evidence suggests that
and pentose phosphate pathways in these tumours109. CRC tumours can also directly transport and metabo-
However, studies in KRAS-​driven PDAC cells and mouse lize fructose from the diet: consumption of moderate
models show that these tumours have increased reliance amounts of fructose enhances intestinal tumour
on glutamine anapleurosis through a non-​canonical enzy- growth in mice by amplifying the expression of hypoxia
matic pathway110. Also, in an apparent exception to the response genes. Fructose’s effect on tumour growth
trend of tumours sharing metabolic preferences with can be negated by targeting the fructolytic enzyme,
their tissue origins, BCAA consumption is decreased in ketohexo­kinase (KHK), or the glycolytic enzyme pyru-
PDACs relative to normal pancreas in mice37,106. In vitro vate kinase isoform M2 (PKM2), which plays a critical
and mouse studies suggest that KRAS-​driven PDAC cells role in mediating hypoxia signalling122,123.
rely on scavenging extracellular amino acids via macro­
pinocytosis, in contrast to KRAS-​driven tumours of the Liver. The pancreas and intestines drain into a pool
lung, as discussed above37,102,111,112. Mouse models and of venous blood that is directly filtered and sampled
epidemiological data also suggest that fatty acid uptake by the liver. This unique anatomy makes the liver a
from the serum is an important feature of PDAC and nutrient-​rich and growth factor-​rich environment ideal
pre-​malignant cells in the pancreas. For example, in vitro for meta­static growth and tumorigenesis. For example,
lipid labelling studies as well as gene expression analysis the liver is exposed to high levels of fructose, lactate
of mouse tumours show that RAS-​transformed pan­ and organic acids from the small intestine, and insulin
creatic cells avidly consume unsaturated fatty acids from from the pancreas, as compared with other tissues25,27,116.
their environment, potentially as a way to circumvent The liver is also hormone-​sensitive, and studies of the
hypoxia-​induced inhibition of stearoyl-​CoA desaturase transcriptional profile of mouse livers over time show
(SCD)113,114. Dietary interventions limiting unsaturated high rates of uptake and production of various metab­
fatty acid availability slow tumour growth in mice bear- olites depending on circulating hormones as well as
ing PDAC allografts, and may be viable therapeutic cell-​intrinsic circadian signals124. For example, stable
Anapleurosis options in humans115. isotope labelling studies in pigs show that the liver takes
Typically referring to the up circu­lating fatty acids (primarily unsaturated), amino
‘refilling’ or ‘fuelling’ of
the tricarboxylic acid (TCA)
Intestine. The intestinal epithelium is exposed to large, acids and lactate, and releases glutamate and ketone
cycle with amino acids to periodic fluctuations in nutrients based on the quantity bodies in response to the low insulin and high glucagon
drive biosynthetic reactions. of food intake and the activity of resident microbiota. state induced by fasting97. The liver is also one of the few

www.nature.com/nrc

0123456789();:
Reviews

Ketosis
tissues capable of generating and releasing glucose via Microbiome
A metabolic state defined gluconeogenesis and is the main source of circulating In addition to the typical macronutrients studied in
by low levels of insulin, high glucose in fasted mice25. the context of other tumours, the intestine and liver
hepatic fatty acid oxidation Hepatocellular carcinoma (HCC) is the most common have unique exposures to dietary, microbial and
and increased levels of
circulating ketone bodies.
primary tumour in the liver, and its development and gastrointestinal-​secretory products that must be consi­
progression are closely related to nutrient intake dered. For example, mouse studies reveal that high-​fat
and systemic metabolism. HCC typically evolves grad- diets can promote microbiota species that increase the
ually from chronically diseased liver tissue such as in amount of tumour-​promoting bile acid species in
the setting of non-​alcoholic fatty liver disease (NAFLD), the intestine133. Additional studies in germ-​free versus
which is estimated to affect up to one-​third of the adult conventionally housed mice reveal that the short-​chain
population globally125. As a result, much attention has fatty acid butyrate, produced by microbial metab­olism
been paid to the systemic and local metabolic altera- of dietary carbohydrate in the intestine, is a major
tions contributing to the development of NAFLD and energy source for differentiated colonocytes134. Although
its progression to HCC. For example, epidemiological butyrate production can be modulated by dietary
studies in the past two decades have shown that obe- interventions, existing mouse studies offer conflicting
sity and the metabolic syndrome confer an increased indications about whether such interventions would
risk of NAFLD and its progression to HCC126. Diets be desirable therapies for CRC, and more studies are
associated with obesity such as those high in both fat needed135,136. Microbial short-​chain fatty acids are also
and fructose also promote HCC in chemically induced important substrates for the liver. For example, recent
mouse models127,128. Mechanistically, de novo lipogenesis tracing studies in mice reveal that dietary fructose serves
(DNL) is purported to play an important role in the pro- as both a signal and a substrate for hepatic lipogenesis,
gression from NAFLD to HCC. The exact sequence of in part through its conversion to acetate by gut-​resident
events linking DNL and HCC is still debated, although microbiota116. Global gene expression profiling of human
one theory posits that increased uptake and genera- HCCs also identified a subset of aggressive tumours that
tion of lipids by hepatocytes contribute to a setting of may rely on gut-​derived acetate for fatty acid synthesis,
chronic liver inflammation that selects for progenitor presenting a promising population for dietary and/or
HCC cells125,129. Indeed, a recent study showed that rats microbial interventions137. Beyond these few examples
with an activating mutation in the de novo lipogenic of microbial-​influenced nutrient availability for tumours,
enzyme, acetyl-​C oA carboxylase (ACC), when fed a there are a plethora of studies available in preclinical
high-fructose diet, exhibited increased hepatic carcino- models reporting associations between the microbiome
genesis as well as tumour cell proliferation. Moreover, and cancer initiation, growth and response to therapy,
an ACC inhibitor effectively reduced tumour prolif- which are covered in excellent reviews elsewhere138.
eration in the rats and in human tumour-​derived cell Because elements of the microbiome are clearly influ-
lines130. One substrate that HCCs use for lipogenesis is enced by diet, such mechanistic associations between
glucose, and similar to other cancers, HCCs generally the microbiome and cancer present attractive targets
upregulate glycolysis, although human imaging studies for dietary interventions in human studies.
show that glucose uptake is highly variable and certain
tumours prefer to use alternative nutrient sources such Systemic effects of dietary therapies
as acetate131,132. Dietary interventions could potentially improve tumour
therapy in several ways. For example, diets can elimi-
Box 1 | Dietary interventions as adjuncts to radiotherapy nate specific nutrients that tumours use as fuel. They
can also potentiate other forms of therapy, such as
One particularly promising application of dietary interventions in cancer is to
enhance the efficacy of other therapies such as radiation. Radiation may induce
radiotherapy (Box 1) and chemotherapy, by depriving
distinct dietary susceptibilities that are absent in untreated tumour cells. Because tumours of escape nutrients and signals. Diets can also
radiation is typically delivered over a set period of time, dietary interventions can act secondarily, modulating the abundance of growth
be confined to the treatment window, potentially allowing for improved compliance factors or altering the systemic immune state to affect
and/or a more aggressive intervention. tumour growth and the antitumour immune response,
One mechanism by which diet could improve radiation outcomes is by limiting the respectively10.
ability of tumour cells to recover from DNA damage. For example, in patient-​derived Dietary interventions come in various forms139. Some
and autochthonous tumour mouse models, a methionine-​restricted diet sensitized focus on content, such as energy (caloric) restriction or
tumour cells to radiation via changes in one-​carbon metabolism. A similar dietary macronutrient manipulation, whereas others are defined
approach was adapted for humans in a controlled clinical study, although the effects
by timing, such as intermittent fasting regimens that
on tumour outcomes have yet to be evaluated229.
Diets that lower circulating glucose have also been investigated as adjuncts to
impart intervals of complete or partial energy restric-
radiation. For example, in mouse models of lung and brain cancer, low glucose cell tion, regardless of meal composition. As these inter-
culture conditions or ketosis-​inducing low-​carbohydrate diets specifically increase the ventions have long been utilized in the fields of obesity
susceptibility of tumour cells to radiation230–232. More recently, in a small, randomized and diabetes, we include a brief review of these stud-
trial of recurrent malignant glioma in humans (ERGO2), a very low carbohydrate diet ies to help identify the potentially beneficial metabolic
(VLCD) combined with intermittent fasting was examined as an adjunct to radiation responses that may occur in patients with cancer. For
therapy. The study did not show a statistically significant difference in progression-​free example, studies in subjects with obesity have shown that
survival or overall survival with the diet, but the 9-​day intervention was well tolerated any dietary intervention with a caloric deficiency will
and post hoc analysis showed that patients who were able to achieve lower levels of activate a neurohormonal response leading to weight
circulating glucose by day 6 had significantly improved survival233.
loss, a decrease in insulin and leptin levels, activation

NATuRE REVIEwS | Cancer

0123456789();:
Reviews

Metabolic dysfunction
of lipolysis in adipose tissue and reduced blood glucose significant weight loss at 2 years with only a 22% dropout
A general term collating variability139. rate and a trend towards improved disease-​free survival
multiple abnormalities in (hazard ratio 0.71, 95% confidence interval 0.41–1.24,
glucose and lipid homeostasis Calorie restriction P = 0.23)157,158. Therefore, we eagerly await the results of
such as dyslipidaemia, obesity,
insulin resistance, glucose
Calorie restriction reduces the total daily energy intake the Breast Cancer WEight Loss Study (BWEL Study),
intolerance, diabetes and while maintaining a well-​b alanced macronutrient a randomized phase III trial evaluating calorie restriction
fatty liver disease. ratio. In most clinical applications, subjects reduce as an adjuvant treatment for early breast cancer159.
their caloric intake by 15–30%. This amount of calorie
restriction lowers body weight, fat mass, insulin, thyroid Fasting mimicking diets
hormones and the metabolic rate in adults with During calorie restriction interventions, the period
overweight140–142. These findings persist at 6 months and of fasting between meals is a commonly ignored con-
2 years of intervention141,143, and also occur in subjects founding variable. Mice undergoing calorie restriction
with obesity with pre-​existing metabolic dysfunction144,145. will consolidate food intake into a single meal leading
In studies of calorie restriction, dropout rates of 30–40% to an intermittent, large influx of calories spaced by long
can be achieved when all foods are provided and par- periods of fasting and hyperactivity160. This adaptation
ticipants are highly motivated146,147. Calorie restriction has led some to question whether it is the calorie restric-
is typically well tolerated with few unexpected adverse tion, per se, or the fasting period that mediates the bene­
events148. fits of calorie restriction in mouse models. In carefully
Since the early 1900s, researchers have observed the controlled studies in mice comparing calorie restriction
beneficial effects of calorie restriction in mouse tumour with fasting interventions that provide a normal amount
models149. In addition to reducing tumour incidence in of calories in one meal per day, fasting results in simi-
mice, calorie restriction slows the progression of can- lar improvements in health and survival, regardless of
cer and the incidence of metastasis150. For example, caloric intake, diet composition and bodyweight161,162.
calorie restriction provides greater protection against In small clinical studies of subjects with cancer under-
established breast and intestinal tumour growth and going chemotherapy, fasting lowered pro-​tumorigenic
metastasis151,152. The mechanism by which calorie restric- hormones, reduced adverse events and improved quality
tion could improve tumour outcomes is pleotropic. of life163,164. Fasting also alters the abundance of periph-
Calorie restriction reduces insulin, which lowers tumour eral immune cells such as monocytes and a subset of
PI3K signalling153. Calorie restriction also promotes dendritic cells, which may have beneficial antitumour
bene­ficial changes in the immune signature151, activation effects165. However, prolonged periods of fasting are hard
of antioxidant pathways154 and beneficial reductions in to maintain166. Therefore, several regimens of intermit-
circulating unsaturated fats115. tent fasting have been designed to mimic the benefits of
There are limited human data describing the effects fasting and improve long-​term adherence167. One such
of calorie restriction as an anticancer intervention148. programme is the fasting mimicking diet (FMD) where
Short-​term studies show that it does not increase grade 3 subjects undergo cycles of consuming calorie-​restricted,
or 4 adverse events during chemotherapy, suggesting cal- low-​carbohydrate, low-​protein diets for four or five
orie restriction is safe155,156. In 2007, a phase III trial was consecutive days each month. For example, FMD may
initiated to test the effect of calorie restriction and exer- consist of a 5-​day, plant-​based diet comprising up to
cise (administered as the Diabetes Prevention Program) 600 calories on day 1 and up to 300 calories on days 2–5
on disease-​free survival and overall survival in postmen- (ref.16). The food is not restricted during the remainder of
opausal women with breast cancer. Unfortunately, the the month, so participants get a long break between FMD
accrual was terminated early owing to loss of funding and cycles. In a randomized crossover study of 100 healthy
only 338 of the planned 2,150 women were enrolled. The subjects, 3 FMD cycles reduced body weight and total
available data were promising, showing a small (~4%) but body fat, lowered blood pressure, and decreased blood
insulin and IGF1 levels as compared with the standard
diet168. The intervention was safe with no grade 3 or 4
Box 2 | Time-​restricted feeding
adverse events and a dropout rate (25%) that was better
Time-​restricted feeding is another fasting-​based approach that is emerging as a than calorie restriction intervention trials. When com-
potential anticancer intervention. Time-​restricted feeding prolongs the daily fasting pared head to head with calorie restriction in a ran­
period to at least 14 h. In subjects without cancer, time-​restricted feeding has produced domized controlled trial of 60 obese women, 2 months
mixed results in terms of weight loss and improvements in metabolic markers234–239. of FMD led to similar weight loss and improvements
Interestingly, the clinical data suggest that the metabolic response depends on the
in markers of insulin resistance and muscle mass,
time of day of the eating window240. Restricting food intake to the middle of the day
(early time-​restricted feeding) may be better than late afternoon or evening periods241.
albeit these were not measured using gold-​standard
Early time-​restricted feeding results in nearly 100% compliance with no adverse methods169. Other forms of intermittent fasting, such as
events240. time-​restricted feeding, are under investigation (Box 2).
Time-​restricted feeding abrogates obesity-​enhanced mammary tumour growth in In mouse models of cancer, cycles of fasting reduce
two orthotopic mouse models in the absence of calorie restriction or weight loss242. tumorigenesis and slow tumour growth170–173. When
Time-​restricted feeding also reduces rates of tumour metastasis in mice242,243. By phar- paired with traditional anticancer therapy, FMD cycles
macologically increasing or decreasing tumour insulin exposure, the authors found that can enhance therapeutic efficacy. For example, fast-
the primary effect of time-​restricted feeding is via a reduction in hyperinsulinemia242. ing cycles enhance gemcitabine efficacy in mice with
To our knowledge, no trials of time-​restricted feeding have been initiated in patients prostate cancer xenografts174, and the FMD strategy
with cancer.
leads to durable remission of breast tumours in mice

www.nature.com/nrc

0123456789();:
Reviews

Preclinical Safety/feasibility Efficacy Clinical practice

Low fat NCT04298086

Calorie restriction NCT02750826

Very low carbohydrate NCT05090358

Fasting mimicking NCT04248998

Amino acids NCT05183295

Fig. 3 | Development progress for dietary interventions for cancer. Several dietary interventions are advancing
well along the clinical development pipeline, with specific example trials highlighted. Based on evidence primarily in
subjects with breast cancer, low-​fat diet (LFD) is often recommended to patients with cancer211–213. Calorie restriction is
safe in subjects with cancer, yet a high dropout rate and poor funding have limited formal efficacy studies157,158. Very low
carbohydrate diet (VLCD) and fasting mimicking diet (FMD) are rapidly advancing through safety and feasibility studies,
and efficacy trials are on the horizon in subjects with cancer16,183. Time-​restricted feeding and diets depleted in specific
amino acids have shown efficacy in mouse models, but limited clinical data are available11.

when combined with anti-​o estrogen therapy and of the lack of carbohydrates and glycaemic load, VLCDs
cyclin-​dependent kinase 4 (CDK4) and CDK6 (hereafter are more effective in improving metabolic parameters
CDK4/6) inhibition, a clinically relevant combination13. associated with glycaemic, weight and lipid controls in
The beneficial effects of FMD are dependent on low patients with high body mass index, especially those
levels of insulin, leptin and IGF1; when these hormones with pre-​existing diabetes, as compared with LFDs187.
are co-​administered to mice receiving FMD, the diet’s The long-​term effects of eating a VLCD have not been
antitumour effects are abolished13. evaluated in large, prospective, randomized controlled
FMD is a promising clinical intervention that is rapi­ clinical trials; however, there is supportive evidence of
dly advancing along the clinical development pipeline safety. In a non-​randomized study of participants with
(Fig. 3). In most cases, FMD is safe and feasible with rates type 2 diabetes over 2 years, an intervention including
of grade 3 or 4 adverse events of 13% and dropout rates of telemedicine, health coaching and a personalized VLCD
24% after 3 FMD cycles in subjects with cancer13,16,175. improved metabolic markers with no reported adverse
The beneficial metabolic effects on insulin, IGF1 and events and a 26% dropout rate188. In patients without
leptin observed in mice appear to be conserved in sub- obesity or metabolic disease, the diet may cause low-
jects with cancer while preserving body weight13,15,16,171,175. grade fatigue, headache, nausea, constipation, hypo-
These changes lower immunosuppressive peripheral glycaemia and acidosis within the first few days to
cells and enhance the intratumour cytotoxic response16. weeks189,190. Low-​grade dehydration, hepatitis, pancrea-
Moreover, FMD is safe in combination with a CDK4/6 titis, hypertriglyceridemia, hyperuricaemia, hypercho-
inhibitor and may improve efficacy13. We eagerly await lesterolaemia, hypomagnesaemia and hyponatraemia
more clinical studies with formal tumour outcomes176. have been reported in older people191.
Despite the potential for adverse events, a syste­
Very low carbohydrate (ketogenic) diets matic review of 13 studies using a VLCD in subjects
Some of the systemic metabolic benefits of calorie with various cancers concluded that this approach is
restriction and fasting can be recapitulated by diets with safe, especially when compared with traditional anti-
altered macronutrient ratios. For example, very low cancer therapy192. For example, Cohen et al. performed
carbo­hydrate diets (VLCDs) or low-​fat diets (LFDs) a 12-​week randomized controlled trial in women with
suppress food intake and alter tumorigenic hormones, ovarian or uterine cancer to test the effects of a VLCD
as compared with standard diets177,178. These diets have intervention on metabolic parameters183. Adherence
been studied in patients with cancer for decades, showing to the diet was excellent (dropout rate 19%), and there
adequate safety, feasibility and, in some cases, anticancer were only low-​grade adverse events including hunger,
efficacy data. constipation, fatigue, muscle cramps, diarrhoea and
The VLCD was clinically described in the 1920s as cold extremities. Subjects displayed selective loss of fat
a means of sustaining the epilepsy-​alleviating effects of mass, retention of lean mass and no change in blood
fasting179,180. In this ‘classic’ form, the VLCD has very lipids182,183,193. Importantly, those on the VLCD had sig-
low carbohydrate intake of ≤15 g per day (<5% of calo- nificantly lower measures of insulin production and
ries), a moderate to low protein intake and enough fat to higher physical function scores193. No effects on tumour
make up the rest of the calorie expenditure181. Typically, outcomes have been reported from this cohort. In other
the fats arise from coconut oil, butter, eggs, avocados, situations, the VLCD is not well tolerated. For example,
cheese and meat. This style of eating has been shown to only 4 of 12 subjects with locally advanced head and neck
promote and maintain very low levels of serum insulin cancer could complete a 5-​week VLCD intervention,
in children with epilepsy, adults with obesity with meta- and several adverse events were noted including grade 2
bolic syndrome and patients with cancer182–186. Because nausea, grade 3 fatigue, grade 4 hyperuricaemia and

NATuRE REVIEwS | Cancer

0123456789();:
Reviews

Table 1 | Potential dietary and pharmacological pairings for future study


Tissue type Tumour or host notable growth Potential dietary Potential pharmacologic Preclinical
biomarkers substrates interventions partners evidence
Breast PIK3CA mutation Glucose VLCD or FMD PI3K–mTOR pathway or ER inhibitor 12,13,69,71,73,83

Triple-​negative Glucose or fatty acids LFD or FMD CDK4/6 or EGFR inhibitor 80,173,226

PHGDH elevation Serine Serine and PHGDH inhibitor 78

glycine-​depleted diet
Endometrial PI3K–mTOR pathway Glucose VLCD PI3K–mTOR pathway inhibitor 6,12,86,87

activation
Prostate MYC pathway activation Fructose or fatty acids LFD, calorie restriction DNL pathway inhibitor 88–92,222

or FMD
Lunga KRAS mutation BCAAs, ketones, fructose Unknown Immunotherapy or chemotherapy 37,102,106,227

or lactate
Pancreasa KRAS mutation Fatty acids Calorie restriction SCD inhibitor or chemotherapy 108,115

or VLCD with high


saturated fatty acids
Intestine WNT pathway activation Glucose or fatty acids LFD DNL inhibitors or chemotherapy 119,123,133

SLC2A5 or KHK Glucose or fructoseb Low-​sugar diet KHK inhibitors, DNL inhibitors 122,123

elevation or chemotherapy
Liver Microbiome alterations Fructose, fatty acids or LFD DNL pathway inhibitor 116,126–128,130–132

short-​chain fatty acids


Brain IDH mutation Glutamine, serine or VLCD or serine and PI3K–mTOR pathway or PHGDH 55,58,59,61,223,228

acetate glycine-​depleted diet inhibitor


BCAA, branched-​chain amino acid; CDK4/6, cyclin-​dependent kinase 4 (CDK4) and CDK6; DNL, de novo lipogenesis; EGFR, epidermal growth factor receptor;
ER, oestrogen receptor; FMD, fasting mimicking diet; IDH, isocitrate dehydrogenase; KHK, ketohexokinase; LFD, low-​fat diet; PHGDH, phosphoglycerate
dehydrogenase; SCD, stearoyl-​CoA desaturase; SLC2A5, solute carrier family 2 member 5; VLCD, very low carbohydrate diet. aHigh risk for weight loss so diets
imposing a calorie restriction should be used with caution. bAlso used as a growth signal.

grade 3 acute pancreatitis194. The VLCD is also poorly lower blood cholesterol and reduced food intake in sub-
tolerated in subjects with lung and pancreatic cancer. For jects without cancer206. In fact, several LFDs are endorsed
example, only 3 of 9 subjects with these cancers could by major medical associations including the Dietary
comply with the diet over 2 years195. Patients with head Approaches to Stop Hypertension (DASH) diet, the US
and neck, lung and pancreas cancers have a high risk of Department of Agriculture food pattern, a vegan diet
developing cachexia196, which may be limiting the ability and the American Heart Association (AHA) diet207–210.
to comply with a VLCD. Unlike the other dietary interventions discussed in
There is growing enthusiasm for the use of the VLCD this Review, LFDs have been rigorously tested in large
as an anticancer intervention. A dietary pattern lower populations of subjects with cancer. The Women’s
in carbohydrates and protein, and higher in fat is asso­ Health Initiative Dietary Modification (WHI DM)
ciated with longer survival time in patients with pancre- trial was a prospective, randomized controlled study
atic cancer115. However, it remains unclear whether these in postmenopausal women designed to examine the
patients will be able to tolerate this dietary pattern given long-​term benefits and risks of a LFD on breast can-
the high predilection for weight loss197. This question cer, CRC and cardiovascular disease203,211. Although no
is currently under investigation198. There are also pro­ long-​term reduction in cancer risk or overall mortality
mising data for use of the VLCD in women with breast was observed212, the LFD intervention lowered the inci-
cancer. In a randomized controlled trial in 80 subjects dence of deaths after breast cancer diagnosis203. This
with breast cancer undergoing chemotherapy, a 12-​week finding was more directly assessed in two randomized
VLCD intervention reduced the tumour diameter as trials: the Women’s Intervention Nutrition Study (WINS)
compared with the controls on a standard diet199. We and the Women’s Healthy Eating and Living (WHEL)
interpret these results with caution as the Response study. Both were randomized trials to test the effects of
Evaluation Criteria in Solid Tumours (RECIST)200 were not a LFD intervention in women with previously treated,
used to measure tumour size and dietary fat intake is asso­ early-​stage breast cancer204,205. Both trials achieved about
ciated with increased risk for breast cancer development a 9% reduction in dietary fat intake between intervention
and recurrence in certain subgroups201–205. Nevertheless, and control groups with about a 30% dropout rate after
these are exciting preliminary data that support the need 3 years213. Although there were some notable differences
Response Evaluation for future studies to better define the specific patient in the populations studied and outcomes reported213, the
Criteria in Solid Tumours subsets that will most benefit from the VLCD. WINS and WHEL data suggest that LFDs have beneficial
(RECIST). A validated effects only in subgroups of patients with breast cancer.
and consistent radiologic Low-​fat diets For example, there was a diet-​induced reduction in distal
methodology to evaluate the
activity and efficacy of new
LFDs restrict fat intake to less than 30% of total calories recurrences among women who did not experience hot
cancer therapeutics in solid per day and emphasize intake of vegetables, fruits and flashes214. This benefit may be due to the diet’s ability
tumours. whole grains. LFDs safely promote weight loss, fat loss, to reduce oestradiol concentrations215,216, which may be

www.nature.com/nrc

0123456789();:
Reviews

One-​carbon metabolism
irrelevant in the age of ER blockade. This question is one-​size-​fits-​all to bespoke interventions for clearly
Referring to both the folate currently under investigation217. denoted cancer subtypes. A similar course could be
and methionine cycles that In regard to prostate cancer, the benefits of a LFD are taken for delivering dietary interventions to support
allow cells to generate less clear. Diets high in fat and saturated fat induce a MYC cancer therapy. We propose targeting dietary interven-
one-​carbon units for the
gene expression signature that independently predicts tions to vulnerable tumour tissue types with distinct his-
biosynthesis of important
anabolic precursors and prostate cancer progression and death218. This finding tological and molecular features (Table 1). For example,
for methylation reactions. has led researchers to study the LFD in several cohorts ER+ breast cancers frequently develop PIK3CA muta-
of men with prostate cancer. Despite good adherence, the tions to evade hormonal therapy, as discussed above.
Eastern Cooperative diet does not change serum prostate-​specific antigen, sex These patients may benefit from combining hormonal
Oncology Group
(ECOG). A standardized
hormones, insulin or IGF1 in this setting219–221. Tumours and PI3K inhibitor therapy with a VLCD, which lowers
clinical scoring algorithm that may evade the effects of a LFD by upregulating fatty insulin and reduces the tumour’s ability to maintain rapid
describes a patient’s level of acid synthesis so clinical efficacy might be improved by nutrient uptake12,13. By contrast, because triple-​negative
functioning in terms of their combining a LFD with inhibitors of this pathway222. breast cancers may rely more heavily on exogenous fatty
ability to care for themself,
acids, a LFD may be a better intervention80. CRC growth
daily activity and physical
ability (walking, working and Other dietary interventions can be promoted by fructose and dietary fats, and thus
so on). There are numerous other means to modify the diet that a plant-​based LFD with no added sugar, which has been
might impede tumour growth. For example, depletion of shown to be feasible in humans224, may be appropriate
Subjective Global specific amino acids such as serine and glycine, cysteine for these patients. Pancreatic cancer cells, meanwhile,
Assessment
(SGA). A clinical scoring
and methionine show promising anticancer efficacy in rely on exogenous unsaturated fatty acids, and thus a
algorithm using information preclinical models (nicely reviewed elsewhere11), but calorie-​restricted or VLCD specifically high in saturated
from a patient interview and there are no clinical data yet. Restricting these amino fats may be effective115.
physical examination that acids in preclinical models depletes tumours of neces- Diet interventions should be paired with synergistic
healthcare providers use
sary precursors for one-​carbon metabolism, and thereby pharmacological therapies because it is unlikely that diet
to determine a person’s
nutritional status. inhi­bits established tumour growth223. Furthermore, the alone will be strong enough to combat tumour progres-
addition of certain sugars, vitamins and amino acids to sion. For example, data in mice indicate that FMDs and
the diet may alter nutrient flux and enhance antican- VLCDs can lead to durable tumour regression when they
cer therapy (recently reviewed in detail10). Early-​phase are paired with CDK4/6 and PI3K inhibitors, respec-
clinical studies are ongoing for many of these approaches. tively12,13. Mouse studies and human genomic analyses
also endorse the combination of amino acid-​depleted
Towards precision nutrition for cancer diets and diets low in unsaturated fats with inhibitors
Precision medicine promises to deliver the right drug of metabolic enzymes such as phosphoglycerate dehy-
to the right patient at the right time. This approach drogenase (PHGDH) and SCD, respectively115,223. The
is helping transform the treatment of cancer from ability of calorie restriction and FMD to synergize
with standard-​of-​care chemotherapy is under active
investigation.
Box 3 | recommendations for dietary intervention studies in cancer
The field of precision nutrition is moving at a rapid
• Provide all meals to support subject adherence. pace with numerous pre-​registered trials on the horizon.
• Carefully engineer meal plans in collaboration with an experienced dietician and We would like to highlight several aspects to consider
openly report these details139. Key pieces of information are often missing from when planning a dietary intervention study in patients
published studies and this deficit limits rigor and reproducibility. Studies should with cancer (Box 3). Of note, the field needs to better
include the following. identify and catalogue diet-​related adverse events. All
-- Food weight and energy density. effective anticancer interventions cause toxicity and
-- Macronutrient and micronutrient content. the field needs to agree on the amount of toxicity that
-- Source of the food products. is acceptable from a dietary intervention. In a recently
-- Fasting duration and timing between fasts.
published study16, Vernieri et al. pre-​specified a 20%
• Define appropriate exclusion criteria focused on key effects of the diet intervention. threshold for severe (grade 3 or 4) adverse events arising
-- Subjects with cachexia, anorexia or moderate-​to-​severe protein-​calorie from a FMD. Given that fasting may help mitigate the
malnutrition may not tolerate diets that impose a large calorie restriction. These adverse effects of chemotherapy, this seems a reason­
patients may be screened for using exclusion criteria such as body mass index
able cut-​off value225. Even with adequate safety, a portion
(BMI) < 20 kg/m2, Eastern Cooperative Oncology Group (ECOG) performance
status ≥3, bioimpedance phase angle <4.5° or Subjective Global Assessment (SGA)
of subjects do not adhere to prescribed diets, even with
category B/C. adequate counselling and full menus provided. There are
• Assume dropout rates of 20–30% of subjects and power appropriately.
numerous reasons for this. There are neurohormonal
pathways that strongly regulate behaviour in the set-
• Collect and report pertinent molecular features of the tumour for each subject.
ting of weight loss, and food carries sentimental value
-- Histological and molecular subtype.
in our cultures, serving as a means to connect with loved
-- Genetic alterations.
ones on special occasions. Investigators should assume
• Catalogue and report the adverse events using standard methods. dropout rates of 20–30% during power calculations if the
• Pre-​plan appropriate pharmacodynamic read-​outs. appro­priate pilot data are not available in subjects with
-- Nutrient or hormone measures in the blood and/or tumour tissue. cancer. Additional studies are needed to find the optimal
-- Appropriate changes in signalling pathways in the tumour. time to commence dietary interventions because some
-- Changes in traditional oncological read-​outs such as progression-​free survival and may only be effective in patients with early-​stage can-
overall survival.
cers; we have limited information in this regard. Lastly,

NATuRE REVIEwS | Cancer

0123456789();:
Reviews

we know very little about the effects of dietary inter- with the potential for a successful outcome. Dietary
ventions on tumour metabolism, signalling and immu- intervention studies have relatively high dropout rates
nity in humans, and this should be investigated using and may impose additional toxicity; however, the promi­
appropriate pharmacodynamic read-​outs. sing preclinical and early clinical data support their
further development. We suggest maximizing the likeli-
Conclusion hood of success by designing studies that test diet–drug
Tumour tissue of origin and genetic driver mutations combination therapies based on the tumour tissue of
contribute to distinct metabolic preferences, which can origin, genetic alterations and the degree of interaction
be targeted with diet during cancer therapy. There is an with systemic hormones and metabolites. Future studies
overall impression by the lay press that ‘healthy eating’ should include appropriate pharmacodynamic read-​outs
will aid in the fight against cancer. Although this senti- that catalogue the effects of each diet on tumour growth,
ment is supported by epidemiological data in the setting metabolism and microenvironment composition, which
of cancer prevention6, it is important to remember that are all understudied in this field.
fighting cancer is a high-​risk, high-​reward endeavour
and the acceptable limits of toxicity need to be balanced Published online xx xx xxxx

1. Song, M. & Giovannucci, E. Preventable incidence and 19. Warburg, O. On the origin of cancer cells. Science 37. Mayers, J. R. et al. Tissue-​of-origin dictates branched-​
mortality of carcinoma associated with lifestyle factors 123, 309–314 (1956). chain amino acid metabolism in mutant Kras-​driven
among white adults in the United States. JAMA Oncol. 20. Vander Heiden, M. G., Cantley, L. C. & Thompson, C. B. cancers. Science 353, 1161–1165 (2016).
2, 1154–1161 (2016). Understanding the Warburg effect: the metabolic 38. Swinnen, J. V., Brusselmans, K. & Verhoeven, G.
2. Parkin, D. M., Boyd, L. & Walker, L. C. The fraction requirements of cell proliferation. Science 324, Increased lipogenesis in cancer cells: new players,
of cancer attributable to lifestyle and environmental 1029–1033 (2009). novel targets. Curr. Opin. Clin. Nutr. Metab. Care 9,
factors in the UK in 2010. Br. J. Cancer 105, 21. Chandel, N. S. Metabolism of proliferating cells. 358–365 (2006).
S77–S81 (2011). Cold Spring Harb. Perspect. Biol. 13, a040618 39. DeBerardinis, R. J. & Thompson, C. B. Cellular
3. Blot, W. J. & Tarone, R. E. Doll and Peto’s quantitative (2021). metabolism and disease: what do metabolic outliers
estimates of cancer risks: holding generally true for 22. Hui, S. et al. Glucose feeds the TCA cycle via teach us? Cell 148, 1132–1144 (2012).
35 years. JNCI 107, djv044 (2015). circulating lactate. Nature 551, 115–118 (2017). 40. Santos, C. R. & Schulze, A. Lipid metabolism in cancer.
4. Barclay, A. W., Flood, V. M., Brand-​Miller, J. C. 23. Faubert, B. et al. Lactate metabolism in human lung FEBS J. 279, 2610–2623 (2012).
& Mitchell, P. Validity of carbohydrate, glycaemic tumors. Cell 171, 358–371.e9 (2017). 41. Sugimoto, M. et al. MMMDB: Mouse Multiple
index and glycaemic load data obtained using a 24. Rabinowitz, J. D. & Enerbäck, S. Lactate: the ugly Tissue Metabolome Database. Nucleic Acids Res. 40,
semi-​quantitative food-​frequency questionnaire. duckling of energy metabolism. Nat. Metab. 2, D809–D814 (2012).
Public Health Nutr. 11, 573–580 (2008). 566–571 (2020). 42. Reznik, E. et al. A landscape of metabolic variation
5. Satija, A., Yu, E., Willett, W. C. & Hu, F. B. 25. Hui, S. et al. Quantitative fluxomics of circulating across tumor types. Cell Syst. 6, 301–313.e3
Understanding nutritional epidemiology and metabolites. Cell Metab. 32, 676–688.e4 (2020). (2018).
its role in policy. Adv. Nutr. 6, 5–18 (2015). This study uses stable isotope tracing to 43. Hoadley, K. A. et al. Multiplatform analysis of
6. Goncalves, M. D., Hopkins, B. D. & Cantley, L. C. quantitatively survey the uptake and monitor 12 cancer types reveals molecular classification within
Dietary fat and sugar in promoting cancer development the fate of infused metabolites in healthy mouse and across tissues of origin. Cell 158, 929–944
and progression. Annu. Rev. Cancer Biol. 3, 255–273 tissues in fasted and fed states. (2014).
(2019). 26. Le, M. T. et al. Effects of high-​fructose corn syrup and 44. Akbani, R. et al. A pan-​cancer proteomic perspective
7. Gallagher, E. J. & LeRoith, D. Hyperinsulinaemia sucrose on the pharmacokinetics of fructose and acute on The Cancer Genome Atlas. Nat. Commun. 5, 3887
in cancer. Nat. Rev. Cancer 20, 629–644 (2020). metabolic and hemodynamic responses in healthy (2014).
8. Hopkins, B. D., Goncalves, M. D. & Cantley, L. C. subjects. Metabolism 61, 641–651 (2012). 45. Karimi, P., Islami, F., Anandasabapathy, S.,
Obesity and cancer mechanisms: cancer metabolism. 27. Jang, C. et al. The small intestine converts dietary Freedman, N. D. & Kamangar, F. Gastric cancer:
J. Clin. Oncol. 34, 4277–4283 (2016). fructose into glucose and organic acids. Cell Metab. descriptive epidemiology, risk factors, screening,
9. World Cancer Research/American Institute for 27, 351–361.e3 (2018). and prevention. Cancer Epidemiol. Biomark. Prev. 23,
Cancer Research. Diet, Nutrition, Physical Activity This study uses stable isotope tracing to 700–713 (2014).
and Cancer: A Global Perspective. Continuous Update quantify the uptake and monitor the fate of 46. Chow, W.-H., Dong, L. M. & Devesa, S. S.
Project Expert Report 2018. dietandcancerreport.org orally delivered sugar in healthy mice over time Epidemiology and risk factors for kidney cancer.
(2018). and notes that the intestine metabolizes most Nat. Rev. Urol. 7, 245–257 (2010).
This report provides a comprehensive analysis of ingested fructose. 47. Simpson, I. A. et al. The facilitative glucose transporter
the global research on lifestyle choices and cancer 28. Carreño, D. et al. Fructose and prostate cancer: GLUT3: 20 years of distinction. Am. J. Physiol.
prevention. toward an integrated view of cancer cell metabolism. Endocrinol. Metab. 295, E242–E253 (2008).
10. Kanarek, N., Petrova, B. & Sabatini, D. M. Dietary Prostate Cancer Prostatic Dis. 22, 49–58 (2019). 48. Ding, J. et al. A metabolome atlas of the aging mouse
modifications for enhanced cancer therapy. Nature 29. Chen, W.-L. et al. Enhanced fructose utilization brain. Nat. Commun. 12, 6021 (2021).
579, 507–517 (2020). mediated by SLC2A5 is a unique metabolic feature 49. Venneti, S. & Thompson, C. B. Metabolic
11. Tajan, M. & Vousden, K. H. Dietary approaches to of acute myeloid leukemia with therapeutic potential. reprogramming in brain tumors. Annu. Rev. Pathol.
cancer therapy. Cancer Cell 37, 767–785 (2020). Cancer Cell 30, 779–791 (2016). Mech. Dis. 12, 515–545 (2017).
12. Hopkins, B. D. et al. Suppression of insulin feedback 30. Raivio, K. O., Kekomäki, M. P. & Mäenpää, P. H. 50. Nehlig, A. Brain uptake and metabolism of ketone
enhances the efficacy of PI3K inhibitors. Nature 560, Depletion of liver adenine nucleotides induced by bodies in animal models. Prostaglandins Leukot.
499–503 (2018). d-​fructose: dose-​dependence and specificity of the Essent. Fat. Acids 70, 265–275 (2004).
13. Caffa, I. et al. Fasting-​mimicking diet and hormone fructose effect. Biochem. Pharmacol. 18, 2615–2624 51. Hawkins, R. A. & Biebuyck, J. F. Ketone bodies are
therapy induce breast cancer regression. Nature 583, (1969). selectively used by individual brain regions. Science
620–624 (2020). 31. Taylor, S. R. et al. Dietary fructose improves intestinal 205, 325–327 (1979).
This study of hormone receptor-​positive mouse cell survival and nutrient absorption. Nature 597, 52. Nehlig, A., Boyet, S. & Pereira de Vasconcelos, A.
models shows that FMD combined with CDK4/6 263–267 (2021). Autoradiographic measurement of local cerebral
inhibitor therapy promotes tumour regression. 32. Bu, P. et al. Aldolase B-​mediated fructose metabolism β-​hydroxybutyrate uptake in the rat during postnatal
14. Maddocks, O. D. K. et al. Modulating the therapeutic drives metabolic reprogramming of colon cancer development. Neuroscience 40, 871–878 (1991).
response of tumours to dietary serine and glycine liver metastasis. Cell Metab. https://doi.org/10.1016/ 53. Schönfeld, P. & Reiser, G. Why does brain metabolism
starvation. Nature 544, 372–376 (2017). J.CMET.2018.04.003 (2018). not favor burning of fatty acids to provide energy?
15. de Groot, S. et al. Fasting mimicking diet as an adjunct 33. Weng, Y. et al. SLC2A5 promotes lung adenocarcinoma Reflections on disadvantages of the use of free fatty
to neoadjuvant chemotherapy for breast cancer in cell growth and metastasis by enhancing fructose acids as fuel for brain. J. Cereb. Blood Flow. Metab.
the multicentre randomized phase 2 DIRECT trial. utilization. Cell Death Discov. 4, 38 (2018). 33, 1493–1499 (2013).
Nat. Commun. 11, 3083 (2020). 34. Chen, W.-L. et al. GLUT5-mediated fructose utilization 54. Maurer, G. D. et al. Differential utilization of ketone
16. Vernieri, C. et al. Fasting-Mimicking Diet Is Safe and drives lung cancer growth by stimulating fatty acid bodies by neurons and glioma cell lines: a rationale
Reshapes Metabolism and Antitumor Immunity in synthesis and AMPK/mTORC1 signaling. JCI Insight 5, for ketogenic diet as experimental glioma therapy.
Patients with Cancer. Cancer Discov. 12, 90–107 (2022). e131596 (2020). BMC Cancer 11, 315 (2011).
This study includes transcriptomics data from the 35. DeBerardinis, R. J. et al. Beyond aerobic glycolysis: 55. Yang, C. et al. Glutamine oxidation maintains the TCA
tumour and describes the changes in peripheral transformed cells can engage in glutamine metabolism cycle and cell survival during impaired mitochondrial
blood cell populations before and after a FMD that exceeds the requirement for protein and pyruvate transport. Mol. Cell 56, 414–424 (2014).
intervention in humans. nucleotide synthesis. Proc. Natl Acad. Sci. USA 104, 56. Schell, J. C. et al. A role for the mitochondrial pyruvate
17. Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: 19345–19350 (2007). carrier as a repressor of the Warburg effect and colon
the next generation. Cell 144, 646–674 (2011). 36. Neinast, M. D. et al. Quantitative analysis of the cancer cell growth. Mol. Cell 56, 400–413 (2014).
18. Chandel, N. S. Glycolysis. Cold Spring Harb. Perspect. whole-​body metabolic fate of branched-​chain amino 57. Yan, H. et al. IDH1 and IDH2 mutations in gliomas.
Biol. 13, a040535 (2021). acids. Cell Metab. 29, 417–429.e4 (2019). N. Engl. J. Med. 360, 765–773 (2009).

www.nature.com/nrc

0123456789();:
Reviews

58. Bi, J. et al. Altered cellular metabolism in gliomas — estrogen in murine and human endometrial stromal 113. Khasawneh, J. et al. Inflammation and mitochondrial
an emerging landscape of actionable co-​dependency cells. Endocrinology 150, 1512–1520 (2009). fatty acid β-​oxidation link obesity to early tumor
targets. Nat. Rev. Cancer 20, 57–70 (2020). 85. Rutanen, E. M. Insulin-​like growth factors in promotion. Proc. Natl Acad. Sci. USA 106,
59. Parker, S. J. & Metallo, C. M. Metabolic consequences endometrial function. Gynecol. Endocrinol. 12, 3354–3359 (2009).
of oncogenic IDH mutations. Pharmacol. Ther. 152, 399–406 (1998). 114. Kamphorst, J. J. et al. Hypoxic and Ras-​transformed
54–62 (2015). 86. Memarzadeh, S. et al. Cell-​autonomous activation of cells support growth by scavenging unsaturated
60. Grassian, A. R. et al. IDH1 mutations alter citric the PI3-kinase pathway initiates endometrial cancer fatty acids from lysophospholipids. Proc. Natl Acad.
acid cycle metabolism and increase dependence on from adult uterine epithelium. Proc. Natl Acad. Sci. USA 110, 8882–8887 (2013).
oxidative mitochondrial metabolism. Cancer Res. 74, Sci. USA 107, 17298–17303 (2010). 115. Lien, E. C. et al. Low glycaemic diets alter lipid
3317–3331 (2014). 87. Cheung, L. W. T. et al. High frequency of PIK3R1 and metabolism to influence tumour growth. Nature
61. Ngo, B. et al. Limited environmental serine and PIK3R2 mutations in endometrial cancer elucidates https://doi.org/10.1038/s41586-021-04049-2
glycine confer brain metastasis sensitivity to PHGDH a novel mechanism for regulation of PTEN protein (2021).
inhibition. Cancer Discov. 10, 1352–1373 (2020). stability. Cancer Discov. 1, 170–185 (2011). This study uses mouse models of KRAS-​mutant
62. Ferraro, G. B. et al. Fatty acid synthesis is required 88. Oyama, N. et al. MicroPET assessment of androgenic PDAC and finds that decreasing tumour access
for breast cancer brain metastasis. Nat. Cancer 2, control of glucose and acetate uptake in the rat to unsaturated fats via diet (calorie restriction
414–428 (2021). prostate and a prostate cancer tumor model. or a high saturated fat ketogenic diet) synergizes
63. Mashimo, T. et al. Acetate is a bioenergetic substrate Nucl. Med. Biol. 29, 783–790 (2002). with inhibition of SCD to slow tumour growth.
for human glioblastoma and brain metastases. Cell 89. Farnsworth, W. E. & Brown, R. Androgen on prostate 116. Zhao, S. et al. Dietary fructose feeds hepatic
159, 1603–1614 (2014). biosynthetic reactions. Endocrinology 68, 978–986 lipogenesis via microbiota-​derived acetate. Nature
64. Anderson, S. M., Rudolph, M. C., McManaman, J. L. (1961). 579, 586–591 (2020).
& Neville, M. C. Key stages in mammary gland 90. Cutruzzolà, F. et al. Glucose metabolism in the This study shows that fructose not only serves
development. Secretory activation in the mammary progression of prostate cancer. Front. Physiol. 8, 97 as a signal acting on the liver directly to induce
gland: it’s not just about milk protein synthesis! Breast (2017). lipogenesis but also as a substrate via its
Cancer Res. 9, 204 (2007). 91. Frenette, G., Thabet, M. & Sullivan, R. Polyol pathway conversion to acetate in intestinal microbiota.
65. Menzies, K. K., Lefèvre, C., Macmillan, K. L. & in human epididymis and semen. J. Androl. 27, 117. Pate, K. T. et al. Wnt signaling directs a metabolic
Nicholas, K. R. Insulin regulates milk protein synthesis 233–239 (2006). program of glycolysis and angiogenesis in colon
at multiple levels in the bovine mammary gland. 92. Szabó, Z. et al. Sorbitol dehydrogenase expression is cancer. EMBO J. 33, 1454–1473 (2014).
Funct. Integr. Genomics 9, 197–217 (2009). regulated by androgens in the human prostate. Oncol. 118. Satoh, K. et al. Global metabolic reprogramming
66. Nommsen-​Rivers, L. A. Does insulin explain the Rep. 23, 1233–1239 (2010). of colorectal cancer occurs at adenoma stage and
relation between maternal obesity and poor lactation 93. Potter, S. R., Epstein, J. I. & Partin, A. W. Seminal is induced by MYC. Proc. Natl Acad. Sci. USA 114,
outcomes? An overview of the literature. Adv. Nutr. 7, vesicle invasion by prostate cancer: prognostic E7697–E7706 (2017).
407–414 (2016). significance and therapeutic implications. Rev. Urol. 2, 119. Brown, R. E., Short, S. P. & Williams, C. S. Colorectal
67. Menzies, K. K. et al. Insulin, a key regulator of 190–195 (2000). cancer and metabolism. Curr. Colorectal Cancer Rep.
hormone responsive milk protein synthesis during 94. Carreño, D. V. et al. Dietary fructose promotes prostate 14, 226–241 (2018).
lactogenesis in murine mammary explants. Funct. cancer growth. Cancer Res. 81, 2824–2832 (2021). 120. Hao, Y. et al. Oncogenic PIK3CA mutations reprogram
Integr. Genomics 10, 87–95 (2010). 95. Watt, M. J. et al. Suppressing fatty acid uptake has glutamine metabolism in colorectal cancer. Nat.
68. Jung, Y., Kim, T. H., Kim, J. Y., Han, S. & An, Y.-S. therapeutic effects in preclinical models of prostate Commun. 7, 11971 (2016).
The effect of sex hormones on normal breast tissue cancer. Sci. Transl. Med. 11, eaau5758 (2019). 121. Yun, J. et al. Glucose deprivation contributes to the
metabolism. Medicine 98, e16306 (2019). 96. Liu, Y. Fatty acid oxidation is a dominant bioenergetic development of KRAS pathway mutations in tumor
69. Campbell, I. G. et al. Mutation of the PIK3CA gene in pathway in prostate cancer. Prostate Cancer Prostatic cells. Science 325, 1555 (2009).
ovarian and breast cancer. Cancer Res. 64, 7678–7681 Dis. 9, 230–234 (2006). 122. Taylor, S. R. et al. Dietary fructose improves intestinal
(2004). 97. Jang, C. et al. Metabolite exchange between cell survival and nutrient absorption. Nature https://
70. Goncalves, M. D., Hopkins, B. D. & Cantley, L. C. mammalian organs quantified in pigs. Cell Metab. 30, doi.org/10.1038/s41586-021-03827-2 (2021).
Phosphatidylinositol 3-kinase, growth disorders, and 594–606.e3 (2019). 123. Goncalves, M. D. et al. High-​fructose corn syrup
cancer. N. Engl. J. Med. 379, 2052–2062 (2018). 98. Cooper, W. A., Lam, D. C. L., O’Toole, S. A. & enhances intestinal tumor growth in mice. Science
71. Miller, T. W. et al. Hyperactivation of Minna, J. D. Molecular biology of lung cancer. 363, 1345–1349 (2019).
phosphatidylinositol-3 kinase promotes escape J. Thorac. Dis. 5, S479–S490 (2013). 124. Vollmers, C. et al. Time of feeding and the intrinsic
from hormone dependence in estrogen receptor-​ 99. Elia, I. et al. Proline metabolism supports metastasis circadian clock drive rhythms in hepatic gene
positive human breast cancer. J. Clin. Invest. 120, formation and could be inhibited to selectively target expression. Proc. Natl Acad. Sci. USA 106,
2406–2413 (2010). metastasizing cancer cells. Nat. Commun. 8, 15267 21453–21458 (2009).
72. Bosch, A. et al. PI3K inhibition results in enhanced (2017). 125. Anstee, Q. M., Reeves, H. L., Kotsiliti, E., Govaere, O. &
estrogen receptor function and dependence in 100. Elia, I. et al. Breast cancer cells rely on environmental Heikenwalder, M. From NASH to HCC: current concepts
hormone receptor-​positive breast cancer. Sci. Transl. pyruvate to shape the metastatic niche. Nature 568, and future challenges. Nat. Rev. Gastroenterol. Hepatol.
Med. 7, 283ra51 (2015). 117–121 (2019). 16, 411–428 (2019).
73. André, F. et al. Alpelisib for PIK3CA-​mutated, hormone 101. Hensley, C. T. et al. Metabolic heterogeneity in human 126. Saran, U., Humar, B., Kolly, P. & Dufour, J.-F.
receptor–positive advanced breast cancer. N. Engl. J. lung tumors. Cell 164, 681–694 (2016). Hepatocellular carcinoma and lifestyles. J. Hepatol.
Med. 380, 1929–1940 (2019). 102. Commisso, C. et al. Macropinocytosis of protein is 64, 203–214 (2016).
74. Elia, I., Schmieder, R., Christen, S. & Fendt, S.-M. an amino acid supply route in Ras-​transformed cells. 127. Tsuchida, T. et al. A simple diet- and chemical-​induced
Organ-​specific cancer metabolism and its potential Nature 497, 633–637 (2013). murine NASH model with rapid progression of
for therapy. Handb. Exp. Pharmacol. 233, 321–353 103. Brunelli, L., Caiola, E., Marabese, M., Broggini, M. steatohepatitis, fibrosis and liver cancer. J. Hepatol.
(2016). & Pastorelli, R. Capturing the metabolomic diversity 69, 385–395 (2018).
75. Deblois, G. & Giguère, V. Oestrogen-​related receptors of KRAS mutants in non-​small-cell lung cancer cells. 128. Healy, M. E. et al. Dietary sugar intake increases liver
in breast cancer: control of cellular metabolism and Oncotarget 5, 4722–4731 (2014). tumor incidence in female mice. Sci. Rep. 6, 22292
beyond. Nat. Rev. Cancer 13, 27–36 (2013). 104. Davidson, S. M. et al. Environment impacts the (2016).
76. Possemato, R. et al. Functional genomics reveal that metabolic dependencies of ras-​driven non-​small 129. Nakagawa, H. et al. Lipid metabolic reprogramming
the serine synthesis pathway is essential in breast cell lung cancer. Cell Metab. 23, 517–528 (2016). in hepatocellular carcinoma. Cancers 10, 447 (2018).
cancer. Nature 476, 346–350 (2011). 105. Faubert, B. & DeBerardinis, R. J. Analyzing tumor 130. Lally, J. S. V. et al. Inhibition of acetyl-​CoA carboxylase
77. Locasale, J. W. et al. Phosphoglycerate dehydrogenase metabolism in vivo. Annu. Rev. Cancer Biol. 1, by phosphorylation or the inhibitor ND-654 suppresses
diverts glycolytic flux and contributes to oncogenesis. 99–117 (2017). lipogenesis and hepatocellular carcinoma. Cell Metab.
Nat. Genet. 43, 869–874 (2011). 106. Davidson, S. M. et al. Direct evidence for cancer-cell- 29, 174–182.e5 (2019).
78. Sullivan, M. R. et al. Increased serine synthesis autonomous extracellular protein catabolism in 131. Ho, C.-L., Yu, S. C. H. & Yeung, D. W. C. 11C-​acetate
provides an advantage for tumors arising in tissues pancreatic tumors. Nat. Med. 23, 235–241 (2017). PET imaging in hepatocellular carcinoma and other
where serine levels are limiting. Cell Metab. 29, 107. Padanad, M. S. et al. Fatty acid oxidation mediated liver masses. J. Nucl. Med. 44, 213–221 (2003).
1410–1421.e4 (2019). by Acyl-​CoA synthetase long chain 3 is required for 132. Jeon, J. Y. et al. Regulation of acetate utilization
79. Marino, N. et al. Upregulation of lipid metabolism mutant KRAS lung tumorigenesis. Cell Rep. 16, by monocarboxylate transporter 1 (MCT1) in
genes in the breast prior to cancer diagnosis. 1614–1628 (2016). hepatocellular carcinoma (HCC). Oncol. Res. 26,
NPJ Breast Cancer 6, 1–13 (2020). 108. Zhang, A. M. Y. et al. Endogenous hyperinsulinemia 71–81 (2018).
80. Monaco, M. E. Fatty acid metabolism in breast cancer contributes to pancreatic cancer development. 133. Jia, W., Xie, G. & Jia, W. Bile acid–microbiota crosstalk
subtypes. Oncotarget 8, 29487–29500 (2017). Cell Metab. 30, 403–404 (2019). in gastrointestinal inflammation and carcinogenesis.
81. Nickel, A. et al. Adipocytes induce distinct gene 109. Ying, H. et al. Oncogenic Kras maintains pancreatic Nat. Rev. Gastroenterol. Hepatol. 15, 111–128 (2018).
expression profiles in mammary tumor cells and tumors through regulation of anabolic glucose 134. Donohoe, D. R. et al. The microbiome and butyrate
enhance inflammatory signaling in invasive breast metabolism. Cell 149, 656–670 (2012). regulate energy metabolism and autophagy in the
cancer cells. Sci. Rep. 8, 9482 (2018). 110. Son, J. et al. Glutamine supports pancreatic cancer mammalian colon. Cell Metab. 13, 517–526 (2011).
82. Balaban, S. et al. Adipocyte lipolysis links obesity to growth through a KRAS-​regulated metabolic pathway. 135. Tilg, H., Adolph, T. E., Gerner, R. R. & Moschen, A. R.
breast cancer growth: adipocyte-​derived fatty acids Nature 496, 101–105 (2013). The intestinal microbiota in colorectal cancer. Cancer
drive breast cancer cell proliferation and migration. 111. Kamphorst, J. J. et al. Human pancreatic cancer Cell 33, 954–964 (2018).
Cancer Metab. 5, 1 (2017). tumors are nutrient poor and tumor cells actively 136. Kaiko, G. E. et al. The colonic crypt protects stem
83. Yager, J. D. & Davidson, N. E. Estrogen carcinogenesis scavenge extracellular protein. Cancer Res. 75, cells from microbiota-​derived metabolites. Cell 165,
in breast cancer. N. Engl. J. Med. 354, 270–282 544–553 (2015). 1708–1720 (2016).
(2006). 112. Sousa, C. M. et al. Pancreatic stellate cells support 137. Björnson, E. et al. Stratification of hepatocellular
84. Frolova, A. et al. Facilitative glucose transporter tumour metabolism through autophagic alanine carcinoma patients based on acetate utilization.
type 1 is differentially regulated by progesterone and secretion. Nature 536, 479–483 (2016). Cell Rep. 13, 2014–2026 (2015).

NATuRE REVIEwS | Cancer

0123456789();:
Reviews

138. Helmink, B. A., Khan, M. A. W., Hermann, A., 162. Pak, H. H. et al. Fasting drives the metabolic, treatment of severe epilepsy. Dev. Med. Child. Neurol.
Gopalakrishnan, V. & Wargo, J. A. The microbiome, molecular and geroprotective effects of a calorie-​ 31, 152–160 (1989).
cancer, and cancer therapy. Nat. Med. 25, 377–388 restricted diet in mice. Nat. Metab. https://doi.org/ 186. Paoli, A., Mancin, L., Giacona, M. C., Bianco, A. &
(2019). 10.1038/s42255-021-00466-9 (2021). Caprio, M. Effects of a ketogenic diet in overweight
139. Goncalves, M. D. & Maddocks, O. D. Engineered diets 163. Bauersfeld, S. P. et al. The effects of short-​term fasting women with polycystic ovary syndrome. J. Transl. Med.
to improve cancer outcomes. Curr. Opin. Biotechnol. on quality of life and tolerance to chemotherapy in 18, 104 (2020).
70, 29–35 (2021). patients with breast and ovarian cancer: a randomized 187. Choi, Y. J., Jeon, S.-M. & Shin, S. Impact of a ketogenic
140. Das, S. K. et al. Low or moderate dietary energy cross-​over pilot study. BMC Cancer 18, 476 (2018). diet on metabolic parameters in patients with obesity
restriction for long-​term weight loss: what works best? 164. Safdie, F. M. et al. Fasting and cancer treatment in or overweight and with or without ype 2 diabetes:
Obesity 17, 2019–2024 (2009). humans: a case series report. Aging 1, 988–1007 a meta-​analysis of randomized controlled trials.
141. Heilbronn, L. K. et al. Effect of 6-mo. calorie restriction (2009). Nutrients 12, E2005 (2020).
on biomarkers of longevity, metabolic adaptation and 165. Jordan, S. et al. Dietary intake regulates the 188. Athinarayanan, S. J. et al. Long-​term effects of a
oxidative stress in overweight subjects. JAMA 295, circulating inflammatory monocyte pool. Cell 178, novel continuous remote care intervention including
1539–1548 (2006). 1102–1114.e17 (2019). nutritional ketosis for the management of type 2
142. Weiss, E. P. & Holloszy, J. O. Improvements in body 166. Trepanowski, J. F. et al. Effect of alternate-​day diabetes: a 2-year non-​randomized clinical trial.
composition, glucose tolerance, and insulin action fasting on weight loss, weight maintenance, and Front. Endocrinol. 10, 348 (2019).
induced by increasing energy expenditure or cardioprotection among metabolically healthy obese 189. Roehl, K. & Sewak, S. L. Practice paper of the
decreasing energy intake. J. Nutr. 137, 1087–1090 adults: a randomized clinical trial. JAMA Intern. Med. academy of nutrition and dietetics: classic and
(2007). 177, 930–938 (2017). modified ketogenic diets for treatment of epilepsy.
143. Kraus, W. E. et al. 2 years of calorie restriction and 167. Nencioni, A., Caffa, I., Cortellino, S. & Longo, V. D. J. Acad. Nutr. Diet. 117, 1279–1292 (2017).
cardiometabolic risk (CALERIE): exploratory outcomes Fasting and cancer: molecular mechanisms and clinical 190. Bostock, E. C. S., Kirkby, K. C., Taylor, B. V. &
of a multicentre, phase 2, randomised controlled trial. application. Nat. Rev. Cancer 18, 707–719 (2018). Hawrelak, J. A. Consumer reports of ‘keto flu’
Lancet Diabetes Endocrinol. 7, 673–683 (2019). 168. Wei, M. et al. Fasting-​mimicking diet and markers/risk associated with the ketogenic diet. Front. Nutr. 7, 20
144. Look AHEAD Research Group. et al. Cardiovascular factors for aging, diabetes, cancer, and cardiovascular (2020).
effects of intensive lifestyle intervention in type 2 disease. Sci. Transl. Med. 9, eaai8700 (2017). 191. Włodarek, D. Role of ketogenic diets in
diabetes. N. Engl. J. Med. 369, 145–154 (2013). 169. Sadeghian, M., Hosseini, S. A., Zare Javid, A., neurodegenerative diseases (Alzheimer’s disease
145. Mudaliar, U. et al. Cardiometabolic risk factor changes Ahmadi Angali, K. & Mashkournia, A. Effect of fasting-​ and Parkinson’s disease). Nutrients 11, E169
observed in diabetes prevention programs in US mimicking diet or continuous energy restriction on (2019).
settings: a systematic review and meta-​analysis. weight loss, body composition, and appetite-​regulating 192. Klement, R. J., Brehm, N. & Sweeney, R. A. Ketogenic
PLoS Med. 13, e1002095 (2016). hormones among metabolically healthy women with diets in medical oncology: a systematic review with
146. Moreira, E. A. M., Most, M., Howard, J. & Ravussin, E. obesity: a randomized controlled, parallel trial. Obes. focus on clinical outcomes. Med. Oncol. 37, 14
Dietary adherence to long-​term controlled feeding Surg. https://doi.org/10.1007/s11695-020-05202-y (2020).
in a calorie-​restriction study in overweight men and (2021). 193. Cohen, C. W., Fontaine, K. R., Arend, R. C.,
women. Nutr. Clin. Pract. 26, 309–315 (2011). 170. Lee, C. et al. Fasting cycles retard growth of tumors Soleymani, T. & Gower, B. A. Favorable effects
147. Dorling, J. L. et al. Changes in body weight, adherence, and sensitize a range of cancer cell types to of a ketogenic diet on physical function, perceived
and appetite during 2 years of calorie restriction: the chemotherapy. Sci. Transl. Med. 4, 124ra27 (2012). energy, and food cravings in women with ovarian or
CALERIE 2 randomized clinical trial. Eur. J. Clin. Nutr. 171. Brandhorst, S. et al. A periodic diet that mimics endometrial cancer: a randomized, controlled trial.
74, 1210–1220 (2020). fasting promotes multi-​system regeneration, enhanced Nutrients 10, 1187 (2018).
148. Shaikh, H. et al. Body weight management in cognitive performance, and healthspan. Cell Metab. 194. Ma, D. C. et al. Ketogenic diet with concurrent
overweight and obese breast cancer survivors. 22, 86–99 (2015). chemoradiation in head and neck squamous cell
Cochrane Database Syst. Rev. 12, CD012110 172. Di Tano, M. et al. Synergistic effect of fasting-​mimicking carcinoma: preclinical and phase 1 trial results.
(2020). diet and vitamin C against KRAS mutated cancers. Radiat. Res. 196, 213–224 (2021).
149. Rous, P. The influence of diet on transplanted and Nat. Commun. 11, 2332 (2020). 195. Zahra, A. et al. Consuming a ketogenic diet while
spontaneous mouse tumors. J. Exp. Med. 20, 173. Salvadori, G. et al. Fasting-​mimicking diet blocks receiving radiation and chemotherapy for locally
433–451 (1914). triple-​negative breast cancer and cancer stem cell advanced lung cancer and pancreatic cancer: the
150. Lv, M., Zhu, X., Wang, H., Wang, F. & Guan, W. Roles escape. Cell Metab. 33, 2247–2259.e6 (2021). University of Iowa experience of two phase 1 clinical
of caloric restriction, ketogenic diet and intermittent 174. D’Aronzo, M. et al. Fasting cycles potentiate the trials. Radiat. Res. 187, 743–754 (2017).
fasting during initiation, progression and metastasis efficacy of gemcitabine treatment in in vitro and 196. Baracos, V. E., Martin, L., Korc, M., Guttridge, D. C. &
of cancer in animal models: a systematic review and in vivo pancreatic cancer models. Oncotarget 6, Fearon, K. C. H. Cancer-​associated cachexia. Nat. Rev.
meta-​analysis. PLoS ONE 9, e115147 (2014). 18545–18557 (2015). Dis. Prim. 4, 17105 (2018).
151. Pomatto-​Watson, L. C. D. et al. Daily caloric restriction 175. Valdemarin, F. et al. Safety and feasibility of fasting-​ 197. Tan-​Shalaby, J. L. et al. Modified Atkins diet in
limits tumor growth more effectively than caloric cycling mimicking diet and effects on nutritional status and advanced malignancies — final results of a safety and
regardless of dietary composition. Nat. Commun. 12, circulating metabolic and inflammatory factors in feasibility trial within the Veterans Affairs Pittsburgh
6201 (2021). cancer patients undergoing active treatment. Cancers Healthcare System. Nutr. Metab. 13, 52 (2016).
152. Castejón, M. et al. Energy restriction and colorectal 13, 4013 (2021). 198. US National Laboratory of Medicine. ClinicalTrials.gov
cancer: a call for additional research. Nutrients 12, 176. Vernieri, C. et al. Exploiting FAsting-​mimicking Diet https://clinicaltrials.gov/ct2/show/NCT04631445
114 (2020). and MEtformin to Improve the Efficacy of Platinum-​ (2022).
153. Kalaany, N. Y. & Sabatini, D. M. Tumours with PI3K pemetrexed Chemotherapy in Advanced LKB1- 199. Khodabakhshi, A. et al. Effects of ketogenic metabolic
activation are resistant to dietary restriction. Nature inactivated Lung Adenocarcinoma: the FAME trial. therapy on patients with breast cancer: a randomized
458, 725–731 (2009). Clin. Lung Cancer 20, e413–e417 (2019). controlled clinical trial. Clin. Nutr. 40, 751–758
154. Pearson, K. J. et al. Nrf2 mediates cancer protection 177. Boden, G., Sargrad, K., Homko, C., Mozzoli, M. (2021).
but not prolongevity induced by caloric restriction. & Stein, T. P. Effect of a low-​carbohydrate diet on 200. Eisenhauer, E. A. et al. New Response Evaluation
Proc. Natl Acad. Sci. USA 105, 2325–2330 (2008). appetite, blood glucose levels, and insulin resistance in Criteria in Solid Tumours: revised RECIST guideline
155. de Man, F. M. et al. Effects of protein and calorie obese patients with type 2 diabetes. Ann. Intern. Med. (version 1.1). Eur. J. Cancer 45, 228–247 (2009).
restriction on the metabolism and toxicity profile of 142, 403–411 (2005). 201. Thiébaut, A. C. M. et al. Dietary fat and
irinotecan in cancer patients. Clin. Pharmacol. Ther. 178. Shintani, T. T., Hughes, C. K., Beckham, S. & postmenopausal invasive breast cancer in the National
109, 1304–1313 (2021). O’Connor, H. K. Obesity and cardiovascular risk Institutes of Health–AARP Diet and Health Study
156. Orgel, E. et al. Caloric and nutrient restriction to intervention through the ad libitum feeding of cohort. J. Natl Cancer Inst. 99, 451–462 (2007).
augment chemotherapy efficacy for acute lymphoblastic traditional Hawaiian diet. Am. J. Clin. Nutr. 53, 202. Howe, G. R., Friedenreich, C. M., Jain, M. & Miller, A. B.
leukemia: the IDEAL trial. Blood Adv. 5, 1853–1861 1647S–1651S (1991). A cohort study of fat intake and risk of breast cancer.
(2021). 179. Wilder, R. M. The effect of ketonemia on the course J. Natl Cancer Inst. 83, 336–340 (1991).
157. Goodwin, P. J. et al. The LISA randomized trial of a of epilepsy. Mayo Clin. Bull. 2, 1 (1921). 203. Chlebowski, R. T. et al. Low-​fat dietary pattern
weight loss intervention in postmenopausal breast 180. Geyelin, H. R. Fasting as a method for treating and breast cancer mortality in the Women’s Health
cancer. NPJ Breast Cancer 6, 6 (2020). epilepsy. M. Rec. 99, 2 (1921). Initiative randomized controlled trial. J. Clin. Oncol.
158. Goodwin, P. J. et al. Randomized trial of a telephone-​ 181. Peterman, M. G. The ketogenic diet in the treatment of 35, 2919–2926 (2017).
based weight loss intervention in postmenopausal epilepsy: a preliminary report. Am. J. Dis. Child. 28, 5 204. Pierce, J. P. et al. Influence of a diet very high in
women with breast cancer receiving letrozole: the LISA (1924). vegetables, fruit, and fiber and low in fat on prognosis
trial. J. Clin. Oncol. 32, 2231–2239 (2014). 182. Cohen, C. W., Fontaine, K. R., Arend, R. C. & following treatment for breast cancer: the Women’s
159. Ligibel, J. A. et al. Randomized phase III trial Gower, B. A. A ketogenic diet is acceptable in women Healthy Eating and Living (WHEL) randomized trial.
evaluating the role of weight loss in adjuvant treatment with ovarian and endometrial cancer and has no JAMA 298, 289–298 (2007).
of overweight and obese women with early breast adverse effects on blood lipids: a randomized, 205. Chlebowski, R. T. et al. Dietary fat reduction and
cancer (Alliance A011401): study design. NPJ Breast controlled trial. Nutr. Cancer 72, 584–594 breast cancer outcome: interim efficacy results from
Cancer 3, 37 (2017). (2020). the Women’s Intervention Nutrition Study. J. Natl
160. Acosta-​Rodríguez, V. A., de Groot, M. H. M., 183. Cohen, C. W. et al. A ketogenic diet reduces central Cancer Inst. 98, 1767–1776 (2006).
Rijo-​Ferreira, F., Green, C. B. & Takahashi, J. S. obesity and serum insulin in women with ovarian 206. Hall, K. D. et al. Effect of a plant-​based, low-​fat diet
Mice under caloric restriction self-​impose a temporal or endometrial cancer. J. Nutr. 148, 1253–1260 versus an animal-​based, ketogenic diet on ad libitum
restriction of food intake as revealed by an automated (2018). energy intake. Nat. Med https://doi.org/10.1038/
feeder system. Cell Metab. 26, 267–277.e2 184. Hyde, P. N. et al. Dietary carbohydrate restriction s41591-020-01209-1 (2021).
(2017). improves metabolic syndrome independent of weight 207. Lichtenstein, A. H. et al. 2021 dietary guidance to
161. Mitchell, S. J. et al. Daily fasting improves health and loss. JCI Insight 4, e128308 (2019). improve cardiovascular health: a scientific statement
survival in male mice independent of diet composition 185. Schwartz, R. M., Boyes, S. & Aynsley-​Green, A. from the American Heart Association. Circulation
and calories. Cell Metab. 29, 221–228.e3 (2019). Metabolic effects of three ketogenic diets in the 144, e472–e487 (2021).

www.nature.com/nrc

0123456789();:
Reviews

208. Evert, A. B. et al. Nutrition therapy recommendations 222. Zadra, G. et al. Inhibition of de novo lipogenesis 238. Chow, L. S. et al. Time-​restricted eating effects on
for the management of adults with diabetes. Diabetes targets androgen receptor signaling in castration-​ body composition and metabolic measures in humans
Care 36, 3821–3842 (2013). resistant prostate cancer. Proc. Natl Acad. Sci. USA who are overweight: a feasibility study. Obesity 28,
209. U.S. Department of Agriculture and U.S. 116, 631–640 (2019). 860–869 (2020).
Department of Health and Human Services. 223. Tajan, M. et al. Serine synthesis pathway inhibition 239. Liu, D. et al. Calorie restriction with or without time-​
Dietary Guidelines for Americans, 2020-2025. cooperates with dietary serine and glycine limitation restricted eating in weight loss. N. Engl. J. Med. 386,
https://www.dietaryguidelines.gov/sites/default/ for cancer therapy. Nat. Commun. 12, 366 (2021). 1495–1504 (2022).
files/2021-03/Dietary_Guidelines_for_Americans- 224. Treasure, M. et al. A pilot study of a low glycemic 240. Sutton, E. F. et al. Early time-​restricted feeding
2020-2025.pdf (2020). load diet in patients with stage I–III colorectal cancer. improves insulin sensitivity, blood pressure, and
210. Sacks, F. M. et al. Soy protein, isoflavones, and J. Gastrointest. Oncol. 12, 910–920 (2021). oxidative stress even without weight loss in men with
cardiovascular health: an American Heart Association 225. Gabel, K., Cares, K., Varady, K., Gadi, V. & Tussing-​ prediabetes. Cell Metab. 27, 1212–1221.e3 (2018).
Science Advisory for professionals from the Nutrition Humphreys, L. Current evidence and directions for 241. Xie, Z. et al. Randomized controlled trial for time-​
Committee. Circulation 113, 1034–1044 (2006). intermittent fasting during cancer chemotherapy. restricted eating in healthy volunteers without obesity.
211. Chlebowski, R. T. et al. Association of low-​fat dietary Adv. Nutr. https://doi.org/10.1093/advances/ Nat. Commun. 13, 1003 (2022).
pattern with breast cancer overall survival: a secondary nmab132 (2021). 242. Das, M. et al. Time-​restricted feeding normalizes
analysis of the Women’s Health Initiative randomized 226. Ni, Y. et al. Death effector domain-​containing protein hyperinsulinemia to inhibit breast cancer in obese
clinical trial. JAMA Oncol. 4, e181212 (2018). induces vulnerability to cell cycle inhibition in triple-​ postmenopausal mouse models. Nat. Commun. 12,
This work shows that in a trial of ~50,000 women negative breast cancer. Nat. Commun. 10, 2860 565 (2021).
with no history of breast cancer, those randomized (2019). 243. Yan, L., Sundaram, S., Mehus, A. A. & Picklo, M. J.
to a LFD who subsequently developed breast 227. McCleland, M. L. et al. Lactate dehydrogenase B Time-​restricted feeding attenuates high-​fat diet-​
cancer had better overall survival than those is required for the growth of KRAS-​dependent lung enhanced spontaneous metastasis of lewis lung
randomized to a control diet who subsequently adenocarcinomas. Clin. Cancer Res. 19, 773–784 carcinoma in mice. Anticancer. Res. 39, 1739–1748
developed breast cancer. (2013). (2019).
212. Thomson, C. A. et al. Cancer incidence and mortality 228. Venneti, S. et al. Glutamine-​based PET imaging
during the intervention and postintervention periods facilitates enhanced metabolic evaluation of gliomas Acknowledgements
of the Women’s Health Initiative Dietary Modification in vivo. Sci. Transl. Med. 7, 274ra17 (2015). This work was supported, in part, by National Institutes of
trial. Cancer Epidemiol. Biomark. Prev. 23, 229. Gao, X. et al. Dietary methionine influences therapy Health (NIH) K08CA230318 (M.D.G.), 2020 AACR–The
2924–2935 (2014). in mouse cancer models and alters human metabolism. Mark Foundation for Cancer Research ‘Science of the Patient’
213. Pierce, J. P. Diet and breast cancer prognosis: making Nature 572, 397–401 (2019). (SOP) Grant Number 20-60-51-​G ONC (M.D.G.), NIH
sense of the WHEL and WINS trials. Curr. Opin. Obstet. 230. Safdie, F. et al. Fasting enhances the response of R35CA197588 (L.C.C.) and a grant from the Breast Cancer
Gynecol. 21, 86–91 (2009). glioma to chemo- and radiotherapy. PLoS ONE 7, Research Foundation (L.C.C.).
214. Gold, E. B. et al. Dietary pattern influences breast e44603 (2012).
cancer prognosis in women without hot flashes: 231. Abdelwahab, M. G. et al. The ketogenic diet is Author contributions
the Women’s Healthy Wating and Living trial. J. Clin. an effective adjuvant to radiation therapy for the S.R.T., J.N.F. and M.D.G. researched data for the article and
Oncol. 27, 352–359 (2009). treatment of malignant glioma. PLoS ONE 7, e36197 wrote the article. All authors contributed substantially to
215. Rose, D. P., Connolly, J. M., Chlebowski, R. T., (2012). discussion of the content, and reviewed and/or edited the
Buzzard, I. M. & Wynder, E. L. The effects of a low-​fat 232. Allen, B. G. et al. Ketogenic diets enhance oxidative manuscript before submission.
dietary intervention and tamoxifen adjuvant therapy stress and radio-​chemo-therapy responses in lung
on the serum estrogen and sex hormone-​binding cancer xenografts. Clin. Cancer Res. 19, 3905–3913 Competing interests
globulin concentrations of postmenopausal breast (2013). L.C.C. is a founder, shareholder and member of the scientific
cancer patients. Breast Cancer Res. Treat. 27, 233. Voss, M. et al. ERGO2: a prospective, randomized advisory board of Agios Pharmaceuticals and a founder and
253–262 (1993). trial of calorie-​restricted ketogenic diet and fasting in former member of the scientific advisory board of Ravenna
216. Rock, C. L. et al. Effects of a high-​fiber, low-​fat diet addition to reirradiation for malignant glioma. Int. J. Pharmaceuticals (previously Petra Pharmaceuticals). These
intervention on serum concentrations of reproductive Radiat. Oncol. Biol. Phys. 108, 987–995 (2020). companies are developing novel therapies for cancer. L.C.C.
steroid hormones in women with a history of breast 234. Peeke, P. M., Greenway, F. L., Billes, S. K., Zhang, D. has received research funding from Ravenna Pharmaceuticals
cancer. J. Clin. Oncol. 22, 2379–2387 (2004). & Fujioka, K. Effect of time restricted eating on body outside the covered work. L.C.C. and M.D.G. are co-​founders
217. US National Laboratory of Medicine. ClinicalTrials.gov weight and fasting glucose in participants with obesity: and shareholders of Faeth Therapeutics, which are developing
https://clinicaltrials.gov/ct2/show/NCT04298086 results of a randomized, controlled, virtual clinical dietary and pharmacologic therapies for cancer. M.D.G. has
(2021). trial. Nutr. Diabetes 11, 6 (2021). received speaking and/or consulting fees from Pfizer Inc.,
218. Labbé, D. P. et al. High-​fat diet fuels prostate 235. de Oliveira Maranhão Pureza, I. R. et al. Effects of time-​ Novartis AG, Petra Pharmaceuticals, Scorpion Therapeutics
cancer progression by rewiring the metabolome and restricted feeding on body weight, body composition and Faeth Therapeutics. M.D.G.’s laboratory has received
amplifying the MYC program. Nat. Commun. 10, and vital signs in low-​income women with obesity: financial support from Pfizer Inc. outside the covered work.
4358 (2019). a 12-month randomized clinical trial. Clin. Nutr. https:// All other authors report no competing interests.
219. Link, L. B., Thompson, S. M., Bosland, M. C. & doi.org/10.1016/j.clnu.2020.06.036 (2020).
Lumey, L. H. Adherence to a low-​fat diet in men with 236. Lowe, D. A. et al. Effects of time-​restricted eating Peer review information
prostate cancer. Urology 64, 970–975 (2004). on weight loss and other metabolic parameters in Nature Reviews Cancer thanks Stephan Herzig, Ömer Yilmaz
220. Aronson, W. J. et al. Phase II prospective randomized women and men with overweight and obesity: the and the other, anonymous, reviewer(s) for their contribution to
trial of a low-​fat diet with fish oil supplementation in treat randomized clinical trial. JAMA Intern. Med. the peer review of this work.
men undergoing radical prostatectomy. Cancer Prev. https://doi.org/10.1001/jamainternmed.2020.4153
Res. 4, 2062–2071 (2011). (2020). Publisher’s note
221. Aronson, W. J. et al. Growth inhibitory effect of low fat 237. Wilkinson, M. J. et al. Ten-​hour time-​restricted eating Springer Nature remains neutral with regard to jurisdictional
diet on prostate cancer cells: results of a prospective, reduces weight, blood pressure, and atherogenic lipids claims in published maps and institutional affiliations.
randomized dietary intervention trial in men with in patients with metabolic syndrome. Cell Metab. 31,
prostate cancer. J. Urol. 183, 345–350 (2010). 92–104.e5 (2020). © Springer Nature Limited 2022

NATuRE REVIEwS | Cancer

0123456789();:

You might also like