You are on page 1of 11

PerSpeCTIVeS

of great importance to many patients with


OPINION
cancer and their families. Diet and nutrition
have long been appreciated to contribute
A framework for examining how diet to cancer risk (not to be confused with
cancer progression, as discussed later)12.
impacts tumour metabolism For example, epidemiological, clinical
and laboratory research have linked
obesity and excess calorie consumption
Evan C. Lien and Matthew G. Vander Heiden    with increased cancer risk, while a low-​
carbohydrate, low-​fat diet is associated
Abstract | The way cancer cells utilize nutrients to support their growth and with lower cancer incidence13–16. While
proliferation is determined by cancer cell-​intrinsic and cancer cell-​extrinsic these studies are provocative, how they
factors, including interactions with the environment. These interactions can inform the best diet for individuals is more
define therapeutic vulnerabilities and impact the effectiveness of cancer therapy. complex. Inconsistencies remain in public
Diet-​mediated changes in whole-​body metabolism and systemic nutrient perception because of conflicting findings
in the scientific literature regarding how
availability can affect the environment that cancer cells are exposed to within dietary factors affect cancer risk and the
tumours, and a better understanding of how diet modulates nutrient availability challenges of studying this complex topic in
and utilization by cancer cells is needed. How diet impacts cancer outcomes is human patients12. Nevertheless, efforts have
also of great interest to patients, yet clear evidence for how diet interacts with been made to propose official guidelines
therapy and impacts tumour growth is lacking. Here we propose an experimental on nutrition for cancer prevention by the
American Cancer Society and the World
framework to probe the connections between diet and cancer metabolism.
Cancer Research Fund/American Institute
We examine how dietary factors may affect tumour growth by altering the access for Cancer Research, which have remained
to and utilization of nutrients by cancer cells. Our growing understanding of fairly consistent for the past 20 years17,18.
how certain cancer types respond to various diets, how diet impacts cancer In contrast to cancer risk, relatively
cell metabolism to mediate these responses and whether dietary interventions little is known about how diet influences
may constitute new therapeutic opportunities will begin to provide guidance on tumour progression when a person already
has cancer and whether certain diets
how best to use diet and nutrition to manage cancer in patients. may maximize survival, either alone or in
combination with other therapies. How to
Defining the different metabolic tissue-​specific contexts3–9. Selecting optimize diet for outcome is a concern for
requirements of proliferating cancer cells patients for treatment with antimetabolite most patients, yet beyond extrapolation
has suggested how cancer cell metabolism chemotherapies based on cancer tissue from data related to cancer risk or specific
might be exploited for better cancer of origin has been successful for decades, mechanisms related to nutrient-​associated
treatment1. Genetic alterations in oncogenes so considering only genotype in selecting hormones such as insulin, evidence is scant
or tumour suppressor genes that promote patients for novel treatments targeting for what patients with specific cancer types
cancer reprogramme cellular metabolism metabolism will not be sufficient in situations should eat. Studying this complex question
in a cell-​autonomous manner, diverting key in which systemic metabolism and the is complicated by the difficulties in finding
metabolites such as glucose and glutamine tissue environment impact the tumours’ suitable experimental models, discrepancies
towards biosynthetic processes that support dependence on the target. Thus, a better between animal and human physiology with
tumorigenesis. This insight has led to an understanding of how interactions between respect to diet and the lack of a clear and
improved understanding of how various cancer cells and their microenvironment consistent approach for testing hypotheses
metabolic pathways are regulated at the and macroenvironment define a tumour’s connecting diet with tumour metabolism
molecular level to facilitate the proliferation metabolic vulnerabilities will be critical for and progression.
and growth of cancer cells. From a developing more effective cancer therapies. Here we explore the idea that dietary
therapeutic perspective, it has also motivated One important environmental effects on tumour progression may be
a search for genotype-​specific metabolic determinant of cancer cell metabolism is mediated in part through changes in
vulnerabilities that can be exploited and has diet, which can affect the availability of the access to and utilization of nutrients
been successful in select contexts2. nutrients within tumours10,11. The study by cancer cells. We aim to provide a
Cancer genetics is not the only of cancer metabolism is often perceived scientific and experimental framework for
determinant of nutrient utilization by by the general public as our understanding approaching and testing hypotheses for how
cancer cells. Cancer cell metabolism of the connections between cancer and diet affects tumour progression and response
is also influenced by metabolic constraints nutrition, and the question of whether to therapy. We draw on observations from
imposed by environmental and certain diets may improve prognosis is various diets that have been explored in the

Nature Reviews | Cancer


Perspectives

literature to illustrate the general concepts whole-​body hormone signalling to affect aspects of animal physiology are distinct
underlying this proposed framework. tumour progression, and ways in which from those of human physiology, valuable
We hope that a systematic approach to these signals impact both cancer risk and information can nevertheless be obtained
probing the relationships between diet tumour growth have been extensively from these preclinical animal models and
and cancer metabolism will lead to better reviewed15,19–25. Another way that dietary refine questions that can then be studied in
scientific guidance for incorporating diet factors can affect tumour growth is through human clinical studies, in which control of
and nutrition into cancer therapy and fill effects on the metabolism of cells within all variables is more difficult.
a critical void for patients in the current tumours. Hormonal signalling alterations
approaches to treat this disease. in response to diet likely impinge on Studying diet–tumour interactions
metabolic processes within cancer cells In this Opinion, we focus primarily on
A conceptual framework to alter proliferation; however, dietary approaches to explore how diet impacts
Dietary factors can affect tumour growth effects on tumour progression may also cancer metabolism and progression in
through a variety of mechanisms that alter be mediated in part through changes in mouse models of cancer so as to review key
cancer cell metabolism. Multiple studies the access to and utilization of nutrients concepts and questions that surround each
have examined how different diets influence by cancer cells. Perturbations to dietary of the four premises in the previous section.
compositions contribute to changes in These premises have been tested in various
plasma metabolite levels, which will in turn forms in the literature, and here we draw on
Diet influence metabolite levels in the tumour observations from various diets to outline
Nutrients Blood microenvironment and thereby alter cancer the evidence for the principles underlying
1 vessel
1 cell metabolism and therapeutic responses this conceptual framework.
(Fig. 1). This hypothesis rests on several
key premises: How does dietary composition affect
tumour growth and progression? There
1. Changes in dietary composition can are extensive data showing that dietary
affect the growth and progression of changes can modulate the growth of various
Tumour tumours. tumour types in animal models. Different
cell
2. The concentrations of metabolites that diets can either inhibit or accelerate tumour
are available to a tumour can be changed growth, and studying both types from
by altering components within the diet. the perspective of understanding cancer
3 Tumour growth 3. Diet-​induced changes in nutrient metabolism is valuable for gaining insight
availability within the tumour into the various physiological metabolic
TIF microenvironment can affect how cancer demands of cancer cells. Low-​carbohydrate
Intracellular Diet-associated cells utilize these nutrients to support diets such as caloric restriction or the
metabolism therapies growth, proliferation and survival. ketogenic diet are generally thought to
4. Diet-​induced changes in cancer cell inhibit the growth of many cancers26.
Fig. 1 | a proposed framework for examining metabolism influence metabolic Caloric restriction, in which overall caloric
how diet impacts tumour metabolism, growth
vulnerabilities that create potential intake is decreased by 30–40% typically
and progression. Diet may affect tumour growth
and progression through mechanisms that alter synergies or antagonism between dietary through carbohydrate limitation, in
cancer cell metabolism. These dietary effects factors and new or existing therapies. particular has been shown across multiple
may be mediated in part through changes in studies to robustly inhibit the growth of
access to and utilization of nutrients by cancer These premises form a framework for diverse tumour types, including breast,
cells within a tumour. We propose a framework understanding the connections between diet lung, prostate, brain, pancreatic and
for exploring the impact of diet on tumour and cancer cell metabolism in a way that is colorectal cancers19,21,26–33. On the other
metabolism and progression that rests on four relevant for patients. In this Opinion, we will hand, a high-​fat diet (HFD), which is
core ideas: (1) the concentrations of metabolites consider diets with different macronutrient distinct from the ketogenic diet because
that are available to a tumour can be changed by compositions (Fig. 2), and the various it still involves substantial carbohydrate
altering components within the diet; (2) diet-​
components of dietary formulations used in consumption, generally increases the
induced changes in nutrient availability within
the tumour environment can affect how cancer animal studies are outlined in Box 1. Factors incidence and growth of tumours, including
cells utilize these nutrients to support growth, that are important when one is designing colorectal, pancreatic, prostate, breast and
proliferation and survival; (3) changes in dietary experiments to study questions related hepatocellular carcinomas23,34–44. Finally,
composition can affect the growth and progres- to diet and cancer can be found in Box 2. specific amino acid-​defined diets have been
sion of tumours; (4) diet-​induced changes in Of note, conducting experiments in patients studied for their ability to inhibit the growth
cancer cell metabolism influence metabolic to probe connections between diet and of various tumour models. For example,
vulnerabilities that create potential synergies or cancer is also relevant, but can be challenging on the basis of observations that serine
antagonism between dietary factors and new because of issues with feasibility, adherence and glycine metabolism is particularly
or existing therapies. Understanding which and the design of appropriate controls12. important for the growth of some cancer
cancer types respond to various diets, how diet
Many of these challenges can be mitigated cells, the serine and glycine-​free diet has
impacts cancer cell metabolism to mediate these
responses and whether dietary interventions may to a certain extent by using animal models been shown to inhibit tumour growth in
open new therapeutic opportunities can help of cancer in which aspects such as dietary several mouse cancer models10,45,46. Similarly,
build a foundation for providing better scientific formulation, caloric intake, tumour genetics, it has been long appreciated that compared
guidance for incorporating diet and nutrition cancer type and procurement of biological with normal cells, cancer cells are
into cancer therapy. TIF, tumour interstitial fluid. samples can be controlled. Although more sensitive to methionine starvation,

www.nature.com/nrc
Perspectives

and a methionine-​restricted diet can inhibit Control diet Caloric restriction Ketogenic diet
the growth of multiple tumour types47–51.
A major challenge in interpreting these
dietary studies, however, is the extent
of variability in the observed effects of a
given diet. For example, although caloric
restriction impairs the growth of most
tumour models, the degree to which
different cancer types respond to caloric
restriction can be variable, and even
different tumour subpopulations within
the same tissue in a mouse model of lung High-fat diet Amino acid-defined diets
cancer can have different sensitivities to the
Control protein component
effects of caloric restriction19,29. Similarly, Amino acid mixture that
the ketogenic diet has received considerable approximates casein composition
popular attention in recent years because it Serine and glycine-free protein
has been shown to inhibit tumour growth in component
some cancer models, particularly in mouse Methionine-restricted protein
models of brain cancer52–57, and yet it can component
20% methionine relative
also accelerate tumour progression in other to control
mouse cancer models20,58,59. The observation
of opposing effects of the same diet on
Carbohydrates Fat Protein Restriction
different tumour types illustrates that
caution is needed in extrapolating these
Fig. 2 | Different diets can be defined by the relative contributions of different macronutrients
studies in animals to provide dietary
to caloric intake. Diets are composed of three macronutrient categories — carbohydrates, fat and
recommendations for cancer patients, protein — that each contributes to caloric intake. Diets also contain components that do not contri­
particularly without a clear understanding bute to caloric intake (not shown), such as vitamins, minerals and fibres (Box 1). Altering the relative
of what mechanistically drives these ratios by which these different macronutrients contribute to caloric intake results in different diets,
differences in tumour response. such as caloric restriction, the ketogenic diet and the high-​fat diet, which each have distinct effects
Some of the variability in response on systemic metabolism and whole-​body physiology. Caloric restriction can involve the long-​term
to diet may be explained by technical restriction of any combination of macronutrients, but is typically achieved through the restriction of
experimental differences in study design, carbohydrates while maintaining equal feeding of protein, fat, vitamins and minerals to induce lower
such as differences across studies in diet blood glucose and insulin levels. The degree of restriction differs across studies, but a commonly used
formulation, the composition of the regimen involves a 30–40% restriction in total calories relative to the control. Both the ketogenic diet
and the high-​fat diet are high in fat, with the key distinction being that the high-​fat diet still involves
comparator control diet, the timing or
substantial carbohydrate consumption, resulting in different physiological effects. Amino acid-​defined
method of diet administration and the diets involve replacing the protein component with purified mixtures of amino acids, allowing the
types of animal cancer models being removal of specific amino acids such as serine, glycine and methionine, which can be done in a way
tested (see Box 2 for a discussion of these that does or does not hold the total amount of nitrogen in the diet constant. The control diet shown
experimental considerations). It is also represents the macronutrient composition of a widely used standard laboratory mouse chow.
probable that dietary effects on tumour
growth are context dependent, since
certain properties of a tumour can affect into cancer cell lines can be sufficient to to tumour responses to diet. It is difficult,
whether it is sensitive to a particular diet. confer resistance to caloric restriction19,29. however, to draw strong conclusions about
Variation in experimental approaches across Genotype can also contribute to tumour which cancer types respond best to a given
different studies can make it difficult to response to the ketogenic diet; for example, diet, since observations are typically spread
uncover these factors, and well-​controlled, the ketogenic diet enhances the growth of across different studies rather than being
systematic evaluations of a given diet across BRAFV600E melanoma. The ketogenic diet, contained within a single well-​controlled
multiple tumour types within the same which restricts both carbohydrates and experiment. To illustrate this, use of the
study may be best suited to define tumour protein to limit access to glucogenic amino ketogenic diet for brain cancers appears
characteristics that may dictate responses acids, leads to ketogenesis primarily in the to be the most promising, although the
to diet. These factors could include tumour liver that converts fatty acids into ketone evidence in support of this diet slowing
genotype, tissue of origin or location of bodies such as β-​hydroxybutyrate and glioma growth is spread out across different
the tumour. acetoacetate, and acetoacetate augments experimental brain cancer models. A few
Several studies have demonstrated that BRAF signalling59,60. Finally, while the serine studies demonstrated that the ketogenic
cancer-​promoting genetic alterations in and glycine-​free diet is effective in increasing diet alone can inhibit tumour growth and
oncogenes or tumour suppressor genes survival in a mouse model of Eµ-​Myc-driven extend survival in various mouse models
can predict tumour responses to various lymphoma and a model of intestinal cancer of glioma and glioblastoma52–54,61,62, while
diets. Cancer cell lines with mutations in driven by Apc loss, it does not inhibit the others show no effect in these models63–65.
the PI3K signalling pathway are resistant growth of Kras-​mutant pancreatic and The ketogenic diet can also inhibit tumour
to the growth inhibitory effects of caloric colorectal tumours10. growth in some other cancer types, such as
restriction, and genetically engineering The cancer cell tissue of origin and/or prostate cancer55,56, pancreatic cancer66 and
an activating PI3K pathway mutation tumour location may also contribute gastric cancer57, although the effect size is

Nature Reviews | Cancer


Perspectives

minimal in some studies. Studies in other to be dependent on the expression of the will be an important step in translating
models report little to no effect, such as fatty acid receptor CD36 in metastasis-​ such knowledge into more well-​informed,
in colorectal, lung and pancreatic cancer initiating cancer cells69. Consistently, a HFD scientifically grounded and potentially
models20,67,68, and in some instances the also enhances metastasis in mouse models ‘personalized’ dietary recommendations
ketogenic diet may even accelerate tumour of prostate cancer driven by Pten and Pml for cancer patients.
growth20,58,59. That brain tumours may be loss70. These data suggest that uptake of
most sensitive to the ketogenic diet suggests dietary fatty acids from a HFD may help Do dietary alterations change metabolite
that some aspect of the brain environment facilitate metastasis. In another study, availability for tumours? The idea that
or lineage could contribute to this response, the synthesis and availability of the amino dietary effects on tumour progression
but more work is needed to determine acid asparagine was found to influence the may be mediated in part through changes
whether this is the case. epithelial–mesenchymal transition in breast in the metabolic environment to which
Differences in nutrient availability across cancer cells, and a diet high in asparagine cancer cells are exposed rests on the premise
tissues may also underlie distinct tumour was sufficient to drive increased metastatic that nutrient availability to cancer cells
responses to diet. For example, breast cancer burden71. These data are consistent in relevant microenvironments can be
cells arising in a mouse model driven by with nutrient availability within tissues modulated by dietary changes. Metabolite
Brca1 and Trp53 loss grow more slowly in influencing the ability of cancers to grow quantification by mass spectrometry and
mammary fat pad tissue that may be even in specific sites, and these types of studies nuclear magnetic resonance spectroscopy
more serine deficient when the animals can help reveal the metabolic requirements has made it feasible to gain a more complete
are consuming a serine and glycine-​free associated with metastasis as well as point understanding of how various diets impact
diet. On the other hand, the growth of to potential dietary interventions that help the metabolome in different biological
these same cancer cells in the more serine-​ limit tumour progression. samples72. Measurements of metabolite
replete pancreatic tissue is less sensitive For the most part, the molecular concentrations in the blood, which are more
to perturbations that affect serine and determinants of whether the growth and straightforward and feasible from a technical
glycine availability46. progression of a tumour will respond to a perspective, have been used as a proxy for
In addition to effects of diet on tumour particular diet are not well understood, and understanding nutrient availability to cancer
growth, how dietary interventions impact systematic efforts will be needed to explore cells within tumours. Indeed, various efforts
other aspects of tumour progression such as this question. As we discuss later, identifying have been made to characterize how nutrient
metastasis is also an important question. the responders versus non-​responders to availability in the blood is altered by both
For example, mice into which oral squamous a dietary intervention can aid in dissecting caloric restriction and short-​term fasting73–79,
cell carcinomas had been implanted which aspects of cancer metabolism dictate the ketogenic diet80–82, the HFD83–86, the
developed more lymph node metastases tumour responses. Moreover, understanding serine and glycine-​free diet10,11,46 and the
when fed a HFD. Increased metastatic the specific characteristics of tumours methionine-​restricted diet51,87–91. Box 3
potential under these conditions was found that define their responses to various diets provides a summary of how these diets
affect the levels of several key metabolites
in the blood.
Box 1 | Compositions of experimental rodent diets
However, blood does not always
experimental animal diets generally fall in two categories: standard chow and purified ingredient represent the local environment to
diets150. Chow diets are made from grains and cereals, containing ingredients such as ground corn, which cancer cells are exposed within
ground oats, alfalfa meal, soybean meal and ground wheat. these plant materials are not well a tumour. Thus, diet-​induced changes
defined, so the actual composition of the same diet can vary from batch to batch. therefore, it can in blood metabolite availability may
be difficult to accurately report the exact composition of these diets in studies, to reproduce the not necessarily be perfectly mirrored
identical effects of these diets across different batches and to modify specific components of
in the local metabolic environment of
these diets to test hypotheses. Rather, purified diets in which each nutrient is clearly defined by
a separate ingredient are more suitable for dietary studies in animal cancer models (for examples
tumours. Regions of tumours differ in
see Fig. 2). Purified animal diets are typically composed of four main macronutrient categories: their degree of vascularization, resulting
carbohydrates, fat, protein, and vitamins and minerals. in gradients of more hypoxic and nutrient-​
Carbohydrates are complex sugars (for example, corn starch and maltodextrin) or simple sugars deprived environments92,93. More recent
(for example, glucose, fructose and sucrose). Fat can be high in saturated fatty acids (for example, efforts to characterize tumour interstitial
lard and milk fat) or high in monounsaturated and/or polyunsaturated fatty acids (for example, fish fluid (TIF), which can be isolated by
oil, vegetable oil, canola oil and soybean oil). Consumption of fats with different fatty acid profiles various methods11,94–96, have found that
can impact the fatty acid composition of tissue lipids and membranes151,152. Given the impact of the metabolite composition of TIF can be
distinct fatty acid species on cancer cell proliferation153–159, dietary fat sources should be controlled distinct from that of plasma11,97–99. While
when different diets are being compared. Protein can be sourced from casein supplemented with
these measurements represent an average
sulfur-​containing amino acids or, if a specific subset of amino acids of interest is to be studied,
from a mixture of individual amino acids at levels that approximate casein or at levels that maintain
level of nutrients found in the tumour
isonitrogenicity. vitamins and minerals can be sourced from rodent-​optimized defined mixes. extracellular fluid and thus fail to capture
Most diets are also supplemented with cellulose as a fibre source and antioxidants such as different subdomains known to exist
tert-​butylhydroquinone. within tumours100, not all nutrients in
since each nutrient group in these purified diets is clearly defined by a separate ingredient, TIF are depleted relative to plasma; some
this allows relatively easy manipulation of dietary components and allows diets under study to metabolites are enriched in TIF, implying
be easily reported and reproduced across different studies. Commercial vendors offer support that the tumour microenvironment may
to assist with diet formulation, and this can be useful to ensure that any dietary modifications are not be as deprived of all nutrients as is
made in a controlled manner. important considerations for designing different diets are further sometimes assumed11. Importantly, TIF
discussed in Box 2.
metabolite concentrations can be modulated

www.nature.com/nrc
Perspectives

Box 2 | experimental design considerations for dietary studies in animal cancer models mechanisms for how dietary changes might
impact cancer progression.
Dietary modifications Another key consideration is that dietary
Formulation of experimental diets should be controlled, such that experimental diets are derived perturbations may induce broad changes
from manipulations of a baseline control diet. a common starting point is the aiN-93G diet or in metabolite availability that extend
the aiN-93M diet, which are nutritionally adequate purified diets for rodents developed by the
beyond the specific metabolite of which
american society for Nutrition160. Outside the intended modifications, the experimental diet
should be identical to the control diet, and three important points must be considered. First, the availability was changed in the diet.
the macronutrients that contain calories have different caloric densities: carbohydrates, proteins For instance, the serine and glycine-​free diet
and fat have 4, 4 and 9 kilocalories per gram, respectively. As a result, a higher-​fat diet will be more changes the levels of other amino acids and
calorically dense than a lower-​fat diet. second, on a dietary change, animals will adjust their food potentially other metabolites that have not
intake to maintain the same caloric consumption, not food weight. Finally, because animals eat yet been measured10,11. Whether changes
for calories, all dietary modifications (other than caloric restriction) require reciprocal changes in the availability of other metabolites also
to at least two components: raising the level of one component necessarily means that the level contribute to the effects of this diet is not
of a second component is lowered (Fig. 2). For example, a high-​fat diet often involves replacing known. Similarly, the methionine-​restricted
carbohydrates with fat, which means that it is also a lower-​carbohydrate diet. Due to the different diet not only lowers blood methionine
caloric densities of these two components, this adjustment cannot be made gram for gram, but
levels but both increases and decreases
rather must be made calorie for calorie. Making calorie-​for-calorie adjustments ensures that
animals consuming diets with different caloric densities will eat the same absolute amounts of the levels of numerous other metabolites51.
all nutrients within the diet other than those that are intentionally changed. For a more in-​depth This concept is particularly relevant to diets
discussion of these concepts, see ref.150 with macronutrient compositions that are
known to impact whole-​body systemic
Caloric intake
although animals tend to maintain their caloric intake across different diets, this may not always
metabolism. A low-​fat, high-​carbohydrate
hold true for all species or strains. it is therefore important to measure caloric consumption diet, for example, does not lower blood
between animals consuming different diets to ensure that different diet groups in fact have similar triglyceride levels but rather increases them
daily caloric consumption, as differences in caloric intake may lead to differences in phenotype that by stimulating hepatic de novo lipogenesis103.
are independent of the source of calories. the effect of diet on body weight may also be important Similarly, a HFD not only raises blood lipid
to consider. For example, the ketogenic diet and caloric restriction typically lead to weight loss, levels but also leads to hyperglycaemia104.
and it may be informative to normalize their effects on tumour growth to the concomitant loss in Dietary fructose does not necessarily lead
body weight33. to high levels of circulating fructose unless
Method of diet administration it is administered at high doses, since at
animal diets are frequently fed ad libitum, in which animals have free access to the food. in most lower doses fructose is mainly cleared in the
cases, this method of administration is sufficient provided that caloric consumption between small intestine by conversion into glucose105.
different cohorts is similar. In situations in which caloric consumption across experimental groups Nevertheless, a high-​fructose diet can also
is not consistent, pair feeding strategies can be considered161. Moreover, diets involving food induce hyperglycaemia, hyperinsulinaemia,
restriction should involve controlled daily feedings of both the control group and the experimental
hyperlipidaemia and insulin resistance106,107.
diet group162.
Low-​carbohydrate diets such as caloric
Timing of diet administration restriction and the ketogenic diet not
if the effects of a diet on tumour initiation are being studied, then diet administration should be only lower blood glucose levels but also
initiated before the establishment of a tumour. if tumour maintenance and progression are being
change the concentrations of many other
examined, then the experimental diets should be initiated after the formation of a palpable and/or
metabolites, such as amino acids, ketones
measurable tumour mass.
and lipids73,75,81. Such broad changes elicited
by dietary perturbations illustrate a major
by diet, although the extent to which plasma concentrations in the blood and TIF are challenge in studying the effects of diet
changes in metabolite concentrations in the millimolar range11, and whether on tumour metabolism. Comprehensively
scale with changes in TIF metabolite these diets decrease glucose levels in the defining how various diets of interest
concentrations differs11. Furthermore, the tumour enough to alter metabolism and alter systemic nutrient availability will be
extent to which various diets alter the TIF slow growth is not known, particularly an important first step in understanding
metabolome across different tumour types given that glucose is often not limiting for how diet might impact tumour growth by
is not known. many cancer cells in vitro when present modulating the access to and utilization of
When biological fluids are being above the micromolar range102. Similarly, metabolites by both cancer cells and stromal
analysed in most metabolomic studies, serine and glycine concentrations in the cells within a tumour.
relative differences in metabolite levels blood are ~150–200 µM under a control
are typically measured in response to an diet, and the serine and glycine-​free diet Do diet-​induced changes in nutrient
experimental perturbation. However, lowers the concentration to ~50–100 µM availability affect tumour metabolism?
it may also be informative in many cases (refs10,46). Culturing cancer cells in these Although some efforts have been made
to measure the absolute concentration lower physiological concentrations of serine to evaluate how dietary alterations
changes of metabolites upon a dietary and glycine in vitro can limit proliferation, change systemic nutrient availability in
alteration. For example, the tumour growth-​ thus providing evidence that the serine and the blood, the challenge is determining
inhibiting effects of low-​carbohydrate glycine-​free diet may in fact be impairing how to mechanistically connect these
diets such as caloric restriction and the tumour growth by starving tumours of these changes to dietary effects on tumour
ketogenic diet are frequently attributed in amino acids46. These examples illustrate how metabolism. One approach is to consider
part to their ability to lower blood glucose absolute quantification of metabolites in how the metabolism of cancer cells can
concentrations54,63,64,101. However, glucose biological samples can provide insight into shift depending on the concentrations of

Nature Reviews | Cancer


Perspectives

Box 3 | Diets can change the levels of metabolites available to a tumour and serum111–114, and others have tried
to mimic the ketogenic diet by culturing
the table provides a summary of how the diets described in this Opinion affect the levels of several cells in the presence of ketones such as
key metabolites in the blood. although some metabolites are robustly affected by specific diets β-hydroxybutyrate and acetoacetate66,101,115,116.
(for example, the ketogenic diet always increases ketone body levels), other metabolites may be Tissue culture media supplemented with
affected variably across different studies. Moreover, it is possible that the levels of some metabolites
lipids and/or fatty acids such as palmitate
may have time-​dependent, complex responses that fluctuate depending on how long a diet has
been administered81,86. as more studies begin to apply improved metabolomics platforms to the have also been used to model the effects
study of different diets, we will increase our understanding of how diet may alter the availability of the HFD23, and media either lacking
of a broader range of metabolites to tumours. or containing low levels of serine and
glycine can mimic effects of the serine
Glucose Insulin Ketone Trigly­ neFa Choles­ amino refs and glycine-​free diet10,45,46. Mimicking
bodies cerides terol acids dietary perturbations in vitro might be
Caloric ↓↓ ↓↓ ↑ Variable Variable ↔ Variable 73,75–78,80
further improved by adjusting additional
restriction aspects of medium compositions on
Ketogenic ↓↓ ↓↓ ↑↑ ↔ ↑ ↑ Variable 20,80,81 the basis of the measured physiological
diet concentrations of metabolites found in the
High-​fat ↑↑ ↑↑ ↔ ↑ ↑ ↑ Variable 80,83–86 plasma or TIF from animals consuming a
diet given diet. If plasma or TIF-​like medium
Serine and ↔ ND ND ND ND ND Serine ↓ 10,46 mimics the effects of a diet on tumour
glycine-​free
Glycine ↓
growth, metabolite dropout or add-​back
diet experiments could then be used to identify
Methionine-​ ↔ ↓ ND ↓ ↓ ↔ Methionine ↓ 51,87–91 the key metabolites modulated by the diet
restricted that are limiting for proliferation or survival.
diet It would also provide an experimental
↑, increased; ↓, decreased; ↔, unchanged; ND, not determined; NeFa, non-​esterified fatty acid. system to conduct nutrient tracing
experiments to understand how various
metabolites in their local environment. dietary effects on metabolite levels in the nutrients are utilized by cancer cells.
This has been highlighted by work exploring tumour microenvironment can change Any conclusions derived from in vitro
the metabolic differences observed in the metabolism of a nutrient the local tissue culture models still need to be
cancer cells growing in vitro versus in vivo. concentration of which is not affected by corroborated in vivo, and a variety of
For example, glutamine is highly consumed the dietary intervention. analyses can help characterize how
by lung cancer cells grown in culture, One approach for studying how diet different diets alter tumour metabolism
where it serves as a major anaplerotic might affect cell metabolism is to mimic the in animal models. One approach is to use
carbon source for the tricarboxylic acid diet-​associated metabolic microenvironment mass spectrometry to obtain metabolomic
cycle108. However, when the same cancer in cell culture. A challenge with preclinical profiles of tumours harvested from animals
cells are grown as a tumour in a mouse, dietary studies in cancer is that such fed with different diets. Although the
glutamine contributes minimally to the experiments typically need to be conducted measurement of steady-​state metabolite
tricarboxylic acid cycle within tumours, in animal models. However, recent data levels provides limited information on the
and tumour growth is not dependent on have suggested that culturing cancer cells activities of metabolic pathways117 and bulk
glutaminase activity3,109. Subsequent work in more physiological tissue culture media tumour measurements are complicated
demonstrated that the high reliance of can recapitulate some metabolic features of by cell-​type heterogeneity, such data may
cancer cells on glutamine in culture is in cancer cells that are observed in vivo6–8,110. point to specific metabolites or classes of
part due to supraphysiological levels of the Therefore, measurement of metabolite metabolites that are perturbed by a given
amino acid cystine (the oxidized dimer concentrations in either plasma or TIF diet that may warrant further investigation.
form of cysteine) found in standard culture under a diet (as discussed in the previous Furthermore, there are network analysis
media, and administering a bolus of cystine section) may provide an opportunity to resources that can correlate metabolite
to mice can indeed increase glutamine mimic the metabolic effects of that diet by pool sizes with the expression of metabolic
incorporation into the tricarboxylic acid constructing a tissue culture medium that genes118,119, which may aid in inferring the
cycle within tumours6. Hence, the levels of resembles the diet-​induced TIF or plasma activities of different metabolic pathways.
cystine in the local environment can alter metabolome. Such an approach could then Together, such analyses may reveal
the extent to which cancer cells metabolize provide an experimentally tractable system potential metabolic pathways that are
glutamine. It is notable that in this case it to assess molecular and mechanistic aspects altered within tumours in response to a
is not environmental glutamine levels that of how diet impacts cancer cell metabolism given diet.
modulate glutamine utilization but rather and proliferation. Another tool to characterize how diets
the local concentrations of a seemingly A few efforts have been made to alter affect tumour metabolism is to trace the
unrelated amino acid, illustrating that broad culture media to mimic a specific diet, fate of isotope-​labelled nutrients within
alterations in the metabolic environment although most have only manipulated tumours and other tissues in vivo. This
can result in unexpected changes in nutrient a subset of components within existing method can provide information about how
utilization by cancer cells. Observations medium formulations. For example, a dietary perturbation influences the relative
such as these are consistent with the fact some studies have tried to mimic caloric utilization of the labelled nutrient and its
that many nutrient transporters function restriction or fasting by culturing cells in incorporation into downstream metabolites.
by metabolite exchange and illustrate how media with low concentrations of glucose However, while in vivo metabolite tracing

www.nature.com/nrc
Perspectives

is a powerful tool, the interpretation of such and impair tumour development, there has Diet
labelling data is complicated particularly been much interest in identifying caloric
within the context of animal physiology. restriction mimetics that can mediate the
For example, normal tissues can rapidly effects of caloric restriction without
consume the labelled nutrient and convert the need for significant food restriction.
it into labelled intermediates that can in Compounds such as resveratrol, metformin
turn supply the label indirectly to cancer and rapamycin have been explored as Tumour metabolism
cells within the tumour120. Tumours are potential mimetics because of their ability
also composed of a heterogenous mixture to induce similar metabolic changes as
Metabolic Diet-induced Metabolic
of non-​cancer and cancer cells121,122, caloric restriction, such as decreases in perturbations metabolic or alterations
which both contribute to the labelling blood glucose, insulin and triglyceride that mediate the synthetic that confer
patterns observed within tumours. Label levels15,124–126. Indeed, rapamycin and dietary growth lethal resistance to
response vulnerabilities dietary effects
incorporation is also often assumed to metformin have been extensively studied
represent net nutrient consumption by for their antitumour effects, although it is
a tumour, but this interpretation can be unclear if the mechanism by which caloric Diet Synergistic Diet-
mimetics diet–drug sensitizing
confounded by exchange flux, wherein restriction inhibits tumour growth is the combinations drug targets
rapid substrate–product interconversion same as that of these compounds15.
by reversible metabolic reactions can lead Alternatively, metabolic pathways that Fig. 3 | Understanding how dietary factors
to the labelling of a metabolite regardless correlate with resistance to the effects of a impact tumour metabolism may help incorpo­
of the net direction of that reaction9,117,123. diet may point to metabolic enzymes that rate dietary interventions into cancer therapy.
Some diets may inhibit tumour growth by per-
Finally, another consideration is that many can be genetically or pharmacologically
turbing specific metabolic pathways that influ-
assumptions for interpretation require manipulated to alter the sensitivity of ence cancer progression. Targeting these
steady-​state labelling, particularly when tumours to that diet (Fig. 3). For instance, metabolic pathways pharmacologically could
different diets may alter the kinetics of Kras-​mutant colorectal tumours are mimic the effects of those diets. For example,
labelled-​nutrient uptake in different resistant to the growth inhibiting effects interventions that mediate the effects of caloric
tissues. Depending on the diet, achieving of a serine and glycine-​free diet because restriction without the need for significant food
steady-​state labelling may be challenging KRAS activation leads to the expression of restriction could improve cancer outcomes.
for some labelled nutrients. For example, higher levels of the de novo serine synthesis Alternatively , adaptive metabolic alterations may
tracing labelled glucose in mice consuming genes, rendering these cancer cells less occur that mediate resistance to dietary effects
a ketogenic diet can be difficult because dependent on exogenous sources of serine by tumours. Targeting these pathways may pro-
vide a means to sensitize tumours to the effects
the diet itself lowers blood glucose levels. and glycine10. To show that the serine and
of certain diets. Finally , although dietary changes
Compared with glucose tracing in mice glycine-​free diet inhibits tumour growth may influence overall patient survival, it is
consuming a normal diet, the administration by starving tumours of serine and glycine, unlikely that any one diet can be used on its own
of labelled glucose must be adjusted for genetically ablating the serine synthesis to reliably induce tumour regression. The maxi-
mice consuming a ketogenic diet to avoid pathway in resistant tumours, such as in mum utility of dietary interventions may lie in
substantially raising blood glucose levels Brca1-null, Trp53-null breast cancer cells, potential synergies when combined with either
during the course of the tracing experiment. sensitizes them to the diet46. Conversely, existing or novel cancer therapies.
Moreover, achieving steady-​state labelling overexpressing the serine synthesis pathway
for some metabolites may not be possible genes in sensitive tumours, such as in breast cells do not express the enzymes necessary
via infusion, because while the body cancer cells lacking the expression of these to metabolize ketones65,115,127–129, and the
has a feedback mechanism to maintain genes, makes them resistant to the effects presence of β-​hydroxybutyrate may be able
euglycaemia and allow steady-​state glucose of the serine and glycine-​free diet46. to suppress proliferation and even cause
labelling, such feedback systems do not Another study demonstrated that cell death in some of these cases66,101,130.
exist for all other metabolites. Despite these dietary supplementation with high-​fructose By contrast, other cancer cells can metabolize
caveats, in vivo metabolite tracing methods corn syrup at amounts that mimic human ketones in culture116,131, raising the question
can provide insight into how diets alter consumption of ~12 ounces of sugar-​ of whether a tumour’s ability to consume
tumour metabolism. sweetened beverages per day enhances ketones might contribute to whether it
Once candidate metabolic pathways the growth of intestinal tumours in an responds to a ketogenic diet. In a comparison
have been identified, determining whether Apc-​deficient mouse model107. Tumours between two cancer cell lines, response to a
these metabolic changes mediate the tumour from mice treated with this diet had ketogenic diet appears to be correlated with
response to a diet can also be challenging. increased levels of long-​chain fatty acids, the expression of the ketone metabolism
One useful approach is to compare tumours suggesting that high-​fructose corn syrup genes BDH1 and OXCT1 (ref.115). Xenograft
that respond to a diet with tumours that activates fatty acid synthesis. Of note, growth of Panc-1 cells, a pancreatic cancer
do not respond. For instance, if a metabolic knockout of the fatty acid synthase gene cell line that lacks BDH1 and OXCT1
pathway that promotes proliferation is (Fasn) in Apc-​deficient intestinal tumours expression, is impaired by a ketogenic diet,
consistently inhibited in all tumours that rendered them insensitive to the growth-​ whereas HeLa cells, which express BDH1 and
have impaired growth in response to a promoting effects of high-​fructose corn OXCT1, are resistant to the diet. Knockdown
diet, then genetically or pharmacologically syrup, indicating that de novo lipogenesis of BDH1 and OXCT1 in HeLa cells sensitizes
targeting enzymes in this pathway may is necessary for the tumour response to them to the ketogenic diet. Therefore,
mimic the effects of that diet (Fig. 3). this diet. expression of ketone metabolism genes may
For example, due to observations that Finally, in the case of the ketogenic diet, be one factor that predicts tumour responses
caloric restriction can extend longevity it has been proposed that some cancer to the ketogenic diet, a conclusion that

Nature Reviews | Cancer


Perspectives

will need to be more systemically explored the effects of existing cancer therapies or how dietary interventions can alter cancer
among more tumour models. novel metabolism-​targeting drugs. cell metabolism in a way that sensitizes
In summary, identifying metabolic A few combinations between different tumours to an existing chemotherapy.
pathways that can be perturbed to either diets and cancer drugs have already been Similarly, multiple studies have
mimic a diet or modulate tumour sensitivity explored. For example, the ketogenic diet demonstrated that fasting regimens and
to dietary effects can enhance understanding augments the efficacy of PI3K inhibitors in low-carbohydrate diets such as caloric
of how dietary perturbations alter cancer acute myeloid leukaemia and pancreatic, restriction and the ketogenic diet can
cell metabolism to affect tumour growth. breast, endometrial and bladder cancer sensitize tumours to the effects of various
mouse models20. PI3K inhibitors block chemotherapies while protecting normal
Can dietary changes reveal metabolic insulin signalling, which subsequently tissues (recently reviewed extensively in
vulnerabilities or drug synergies? Although prevents glucose uptake in tissues and leads to ref.136). The methionine-​restricted diet
dietary perturbations can affect tumour an increase in blood glucose concentrations. has also been shown in multiple studies to
progression and may extend survival10,19,33,46, This transient hyperglycaemia induces the synergize with various chemotherapies51,137,138.
few diets, if any, have been shown to release of insulin from the pancreas, which Some studies advocate a short-term but
robustly cause tumour regression on is then proposed to override the effects more extreme dietary change, such as
their own, possibly because restricting of PI3K inhibition in tumours by reactivating extreme caloric restriction (greater than 90%)
nutrients through dietary means typically PI3K signalling. Since the ketogenic diet or fasting for a few days before chemotherapy,
impairs proliferation without affecting lowers blood glucose and insulin levels, immunotherapy or treatment with other
cell survival. An important opportunity it can blunt the rise in blood glucose and targeted agents. These short-term dietary
afforded by dietary interventions may insulin levels caused by PI3K inhibitors, interventions may still enhance the anticancer
therefore lie in potential synergies with thereby improving the antitumour activity effects of therapies and enable patients to
other cancer therapies (Fig. 3). The idea of the drugs. This observation illustrates benefit from a diet without the negative
that diet-​induced changes in cancer cell how the effects of a diet on whole-​body effects associated with long-​term exposure
metabolism may contribute to potential metabolism can be exploited to augment to a diet111,139. These findings have motivated
diet–drug synergies is supported by the efficacy of a targeted therapy currently clinical trials evaluating these diets in
observations that environmental nutrient under development. Moreover, given that combination with chemotherapy140–146.
availability can not only change cancer the ketogenic diet leads to broad changes The mechanisms underlying these
cell metabolism but can also significantly in systemic nutrient availability80,81 and the combinations have largely been attributed
impact the efficacy of existing therapies. PI3K signalling pathway is a major regulator to how these diets lower blood glucose
For example, as described earlier, in of cancer cell metabolism133, there may levels and reduce IGF1 and insulin
the presence of physiological levels of be additional mechanisms by which the signalling25,111,136,139,147. However, these diets
exogenous cystine, lung tumours depend ketogenic diet sensitizes tumours to PI3K may also alter cancer and normal tissue cell
less on glutaminolysis for proliferation, inhibitors by affecting cancer metabolism metabolism in other ways that contribute
which may explain in part why CB-839, or by altering the metabolic environment to increased drug sensitivity. For example,
a glutaminase inhibitor currently in clinical within the tumour. lung and ovarian cancer cell lines that are
trials, may show limited efficacy in some In another recent study, a CRISPR–Cas9 resistant to cisplatin are more sensitive
cancer patients6,132. Whether increasing screen identified histidine metabolism as a to nutrient deprivation and glutamine
cystine availability within tumours in turn modulator of cancer cell sensitivity to the starvation, which leads to the impairment
sensitizes them to glutaminase inhibitors chemotherapeutic drug methotrexate134. Loss of nucleotide synthesis and can resensitize
remains to be determined. Similarly, the of formimidoyltransferase cyclodeaminase these cells to cisplatin-​induced cell death148.
antidiabetic drug metformin can slow (FTCD), which is required for histidine Thus, it is possible that limiting tumour
the growth of lung tumour xenografts in degradation, caused various haematopoietic access to glutamine by fasting regimens in
a mouse model but not in tissue culture cancer cell lines to become more resistant this context could synergize with cisplatin
models, in which supraphysiological to methotrexate. One mechanism for treatment. Further studies will improve our
levels of pyruvate abolish the sensitivity the anticancer effects of methotrexate is the understanding of whether changes in tumour
of cancer cells to metformin4. Another inhibition of dihydrofolate reductase, which metabolism mediated by fasting regimens or
study demonstrated that when leukaemia generates the cofactor tetrahydrofolate low-​carbohydrate diets can contribute in part
and colorectal cancer cells are cultured in (THF)135. Depletion of THF by methotrexate to increased chemotherapy sensitivity.
media with polar metabolite concentrations can lead to cell death because it is required Finally, in some instances certain diets
similar to that of human plasma, they are for various metabolic processes, perhaps will not modulate or even antagonize the
less sensitive to 5-fluorouracil, a commonly most notably nucleotide synthesis. In effects of a specific drug. For instance, even
used chemotherapeutic compound7. the histidine degradation pathway, the though serine and glycine starvation can
Observations such as these demonstrate that FTCD reaction utilizes THF, and histidine synergize with mitochondrial inhibitors
at least in vitro the metabolic environment catabolism was therefore proposed to such as oligomycin and metformin in
of cancer cells can alter their responses to synergize with methotrexate by further some contexts45,149, in other contexts this
various drugs. Extension of this concept depleting the THF pool. Remarkably, combination has no effect or may even
to an in vivo setting implies that diet-​ dietary supplementation with histidine in increase tumour burden10. Taken together,
mediated changes to systemic nutrient tumour-​bearing mice raised histidine levels these examples highlight the value of
availability can also affect how tumours within the tumours and led to increased studying diet and drug combinations
respond to certain cancer therapies. Thus, histidine breakdown, and this synergized in defined contexts. As we increase our
it may be possible to rationally determine with methotrexate treatment to inhibit understanding of how diet-​mediated
whether diet will synergize or antagonize tumour growth. This observation illustrates changes to cancer cell metabolism may

www.nature.com/nrc
Perspectives

reveal new therapeutic strategies, new 9. Muir, A., Danai, L. V. & Vander Heiden, M. G. 32. Shelton, L. M., Huysentruyt, L. C., Mukherjee, P. &
Microenvironmental regulation of cancer Cell Metab. Seyfried, T. N. Calorie restriction as an anti-​invasive
insight will be gained to aid efforts to tailor olism: implications for experimental design and therapy for malignant brain cancer in the VM mouse.
dietary recommendations to specific cancer translational studies. Dis Model. Mech 11, (2018). ASN Neuro. 2, e00038 (2010).
10. Maddocks, O. D. K. et al. Modulating the therapeutic 33. Danai, L. V. et al. Altered exocrine function can
drugs and tumour types. response of tumours to dietary serine and glycine drive adipose wasting in early pancreatic cancer.
starvation. Nature 544, 372–376 (2017). Nature 558, 600–604 (2018).
11. Sullivan, M. R. et al. Quantification of 34. Algire, C., Amrein, L., Zakikhani, M., Panasci, L. &
Conclusion microenvironmental metabolites in murine cancers Pollak, M. Metformin blocks the stimulative effect of
How diet and nutrition affect cancer reveals determinants of tumor nutrient availability. a high-​energy diet on colon carcinoma growth in vivo
Elife 8, e44235 (2019). and is associated with reduced expression of fatty acid
progression is a question of critical 12. Mayne, S. T., Playdon, M. C. & Rock, C. L. synthase. Endocr. Relat. Cancer 17, 351–360 (2010).
importance to many cancer patients, and one Diet, nutrition, and cancer: past, present and future. 35. O’Neill, A. M. et al. High-​fat Western diet-​induced
Nat. Rev. Clin. Oncol. 13, 504–515 (2016). obesity contributes to increased tumor growth in
that will benefit from more study and 13. Khandekar, M. J., Cohen, P. & Spiegelman, B. M. mouse models of human colon cancer. Nutr. Res. 36,
evidence-​based conclusions. It has become Molecular mechanisms of cancer development in 1325–1334 (2016).
obesity. Nat. Rev. Cancer 11, 886–895 (2011). 36. Schulz, M. D. et al. High-​fat-diet-​mediated dysbiosis
increasingly clear that the way cancer cells 14. Kitahara, C. M. et al. Prospective investigation of body promotes intestinal carcinogenesis independently
utilize nutrients to support their growth mass index, colorectal adenoma, and colorectal cancer of obesity. Nature 514, 508–512 (2014).
in the prostate, lung, colorectal, and ovarian cancer 37. Chang, H. H. et al. Incidence of pancreatic cancer
and proliferation is determined not only by screening trial. J. Clin. Oncol. 31, 2450–2459 is dramatically increased by a high fat, high calorie
cancer-​promoting genetic alterations but (2013). diet in KrasG12D mice. PLOS ONE 12, e0184455
15. Meynet, O. & Ricci, J.-E. Caloric restriction and cancer: (2017).
also by the tumour’s interactions with its molecular mechanisms and clinical implications. 38. Nadella, S. et al. Dietary fat stimulates pancreatic
local metabolic environment. Applying this Trends Mol. Med. 20, 419–427 (2014). cancer growth and promotes fibrosis of the tumor
16. Michels, K. B. & Ekbom, A. Caloric restriction and microenvironment through the cholecystokinin
understanding to study the connections incidence of breast cancer. JAMA 291, 1226–1230 receptor. Am. J. Physiol. Gastrointest Liver Physiol.
between diet and cancer provides a (2004). 315, G699–G712 (2018).
17. World Cancer Research Fund/American Institute for 39. Okumura, T. et al. Extra-​pancreatic invasion
framework for how to probe dietary effects Cancer Research. Food, nutrition, physical activity, induces lipolytic and fibrotic changes in the adipose
on nutrient access and utilization by cancer and the prevention of cancer: a global perspective. microenvironment, with released fatty acids enhancing
American Institute for Cancer Research http://www. the invasiveness of pancreatic cancer cells. Oncotarget
cells that influence tumour progression. aicr.org/assets/docs/pdf/reports/Second_Expert_ 8, 18280–18295 (2017).
This could lead to a better understanding of Report.pdf (2007). 40. Philip, B. et al. A high-​fat diet activates oncogenic
18. Kushi, L. H. et al. American cancer society guidelines Kras and COX2 to induce development of pancreatic
which cancer types respond to various diets, on nutrition and physical activity for cancer prevention: ductal adenocarcinoma in mice. Gastroenterology
how diet impacts cancer cell metabolism reducing the risk of cancer with healthy food choices 145, 1449–1458 (2013).
and physical activity. CA Cancer J. Clin. 62, 30–67 41. Huang, M. et al. Diet-​induced alteration of fatty acid
to mediate these responses and whether (2012). synthase in prostate cancer progression. Oncogenesis
dietary interventions may open new 19. Kalaany, N. Y. & Sabatini, D. M. Tumours with 5, e195 (2016).
PI3K activation are resistant to dietary restriction. 42. Kim, S. et al. Myristoylation of Src kinase mediates
therapeutic opportunities in combination Nature 458, 725–731 (2009). Src-​induced and high-​fat diet-​accelerated prostate
with current cancer treatments. It will also 20. Hopkins, B. D. et al. Suppression of insulin feedback tumor progression in mice. J. Biol. Chem. 292,
enhances the efficacy of PI3K inhibitors. Nature 560, 18422–18433 (2017).
build a foundation for providing better 499–503 (2018). 43. Sundaram, S. & Yan, L. High-​fat diet enhances
scientific guidance for incorporating diet 21. Nogueira, L. M. et al. Dose-​dependent effects of mammary tumorigenesis and pulmonary metastasis
and nutrition into cancer therapy. calorie restriction on gene expression, metabolism,
and tumor progression are partially mediated by
and alters inflammatory and angiogenic profiles in
MMTV-​PyMT Mice. AntiCancer Res. 36, 6279–6287
Evan C. Lien1,2 and Matthew G. Vander Heiden   1,2,3* insulin-​like growth factor-1. Cancer Med. 1, 275–288 (2016).
(2012). 44. Ip, B. C., Liu, C., Smith, D. E., Ausman, L. M. &
Koch Institute for Integrative Cancer Research,
1
22. Lu, Z. et al. Fasting selectively blocks development Wang, X. D. High-​refined-carbohydrate and high-​fat
Massachusetts Institute of Technology, Cambridge, of acute lymphoblastic leukemia via leptin-​receptor diets induce comparable hepatic tumorigenesis in
upregulation. Nat. Med. 23, 79–90 (2017). male mice. J. Nutr. 144, 647–653 (2014).
MA, USA.
23. Beyaz, S. et al. High-​fat diet enhances stemness and 45. Maddocks, O. D. et al. Serine starvation induces stress
2
Department of Biology, Massachusetts Institute tumorigenicity of intestinal progenitors. Nature 531, and p53-dependent metabolic remodelling in cancer
of Technology, Cambridge, MA, USA. 53–58 (2016). cells. Nature 493, 542–546 (2013).
24. Hursting, S. D., Dunlap, S. M., Ford, N. A., 46. Sullivan, M. R. et al. Increased serine synthesis
3
Department of Medical Oncology, Dana-​Farber Hursting, M. J. & Lashinger, L. M. Calorie restriction provides an advantage for tumors arising in tissues
Cancer Institute, Boston, MA, USA. and cancer prevention: a mechanistic perspective. where serine levels are limiting. Cell Metab. 29,
Cancer Metab. 1, 10 (2013). 1410–1421 (2019).
*e-​mail: mvh@mit.edu 25. Lee, C. & Longo, V. D. Fasting vs dietary restriction in 47. Breillout, F., Hadida, F., Echinard-​Garin, P., Lascaux, V.
https://doi.org/10.1038/s41568-019-0198-5 cellular protection and cancer treatment: from model & Poupon, M. F. Decreased rat rhabdomyosarcoma
organisms to patients. Oncogene 30, 3305–3316 pulmonary metastases in response to a low methionine
Published online xx xx xxxx (2011). diet. AntiCancer Res. 7, 861–867 (1987).
26. Lv, M., Zhu, X., Wang, H., Wang, F. & Guan, W. 48. Guo, H. et al. Therapeutic tumor-​specific cell cycle
1. Vander Heiden, M. G. & DeBerardinis, R. J. Roles of caloric restriction, ketogenic diet and block induced by methionine starvation in vivo.
Understanding the intersections between metabolism intermittent fasting during initiation, progression and Cancer Res. 53, 5676–5679 (1993).
and cancer biology. Cell 168, 657–669 (2017). metastasis of cancer in animal models: a systematic 49. Hoshiya, Y. et al. Human tumors are methionine
2. Luengo, A., Gui, D. Y. & Vander Heiden, M. G. review and meta-​analysis. PLOS ONE 9, e115147 dependent in vivo. AntiCancer Res. 15, 717–718
Targeting metabolism for cancer therapy. Cell Chem. (2014). (1995).
Biol. 24, 1161–1180 (2017). 27. De Lorenzo, M. S. et al. Caloric restriction reduces 50. Komninou, D., Leutzinger, Y., Reddy, B. S. &
3. Davidson, S. M. et al. Environment impacts the growth of mammary tumors and metastases. Richie, J. P. J. Methionine restriction inhibits colon
metabolic dependencies of Ras-​driven non-​small cell Carcinogenesis 32, 1381–1387 (2011). carcinogenesis. Nutr. Cancer 54, 202–208 (2006).
lung cancer. Cell Metab. 23, 517–528 (2016). 28. Nogueira, L. M., Dunlap, S. M., Ford, N. A. & 51. Gao, X. et al. Dietary methionine influences therapy
4. Gui, D. Y. et al. Environment dictates dependence on Hursting, S. D. Calorie restriction and rapamycin in mouse cancer models and alters human metabolism.
mitochondrial complex I for NAD+ and aspartate inhibit MMTV-​Wnt-1 mammary tumor growth in a Nature https://doi.org/10.1038/s41586-019-1437-3
production and determines cancer cell sensitivity to mouse model of postmenopausal obesity. Endocr. (2019).
metformin. Cell Metab. 24, 716–727 (2016). Relat. Cancer 19, 57–68 (2012). 52. Stafford, P. et al. The ketogenic diet reverses gene
5. Mayers, J. R. et al. Tissue of origin dictates 29. Curry, N. L. et al. Pten-​null tumors cohabiting the expression patterns and reduces reactive oxygen
branched-​chain amino acid metabolism in mutant same lung display differential AKT activation and species levels when used as an adjuvant therapy
Kras-​driven cancers. Science 353, 1161–1165 sensitivity to dietary restriction. Cancer Discov. 3, for glioma. Nutr. Metab. (Lond) 7, 74 (2010).
(2016). 908–921 (2013). 53. Abdelwahab, M. G. et al. The ketogenic diet is
6. Muir, A. et al. Environmental cystine drives glutamine 30. Lanza-​Jacoby, S. et al. Calorie restriction delays an effective adjuvant to radiation therapy for the
anaplerosis and sensitizes cancer cells to glutaminase the progression of lesions to pancreatic cancer treatment of malignant glioma. PLOS ONE 7,
inhibition. Elife 6, e27713 (2017). in the LSL-​KrasG12D; Pdx-1/Cre mouse model of e36197 (2012).
7. Cantor, J. R. et al. Physiologic medium rewires cellular pancreatic cancer. Exp. Biol. Med. (Maywood) 238, 54. Poff, A. M., Ari, C., Seyfried, T. N. & D’Agostino, D. P.
metabolism and reveals uric acid as an endogenous 787–797 (2013). The ketogenic diet and hyperbaric oxygen therapy
inhibitor of ump synthase. Cell 169, 258–272.e17 31. Mukherjee, P., Abate, L. E. & Seyfried, T. N. prolong survival in mice with systemic metastatic
(2017). Antiangiogenic and proapoptotic effects of dietary cancer. PLOS ONE 8, e65522 (2013).
8. Vande Voorde, J. et al. Improving the metabolic fidelity restriction on experimental mouse and human 55. Freedland, S. J. et al. Carbohydrate restriction,
of cancer models with a physiological cell culture brain tumors. Clin. Cancer Res. 10, 5622–5629 prostate cancer growth, and the insulin-​like growth
medium. Sci. Adv. 5, eaau7314 (2019). (2004). factor axis. Prostate 68, 11–19 (2008).

Nature Reviews | Cancer


Perspectives

56. Mavropoulos, J. C. et al. The effects of varying dietary 82. Heischmann, S. et al. Regulation of kynurenine diabetes mediated by hepatic/pancreatic endoplasmic
carbohydrate and fat content on survival in a murine metabolism by a ketogenic diet. J. Lipid Res. 59, reticulum (ER) stress. Mol. Cell Biochem. 423, 93–104
LNCaP prostate cancer xenograft model. Cancer Prev. 958–966 (2018). (2016).
Res. (Phila) 2, 557–565 (2009). 83. Fujisaka, S. et al. Diet, genetics, and the gut microbiome 107. Goncalves, M. D. et al. High-​fructose corn syrup
57. Otto, C. et al. Growth of human gastric cancer cells in drive dynamic changes in plasma metabolites. Cell Rep. enhances intestinal tumor growth in mice. Science
nude mice is delayed by a ketogenic diet supplemented 22, 3072–3086 (2018). 363, 1345–1349 (2019).
with omega-3 fatty acids and medium-​chain triglycerides. 84. Wang, W. et al. Effects of high-​fat diet on plasma profiles 108. Hensley, C. T., Wasti, A. T. & DeBerardinis, R. J.
BMC Cancer 8, 122 (2008). of eicosanoid metabolites in mice. Prostaglandins Other Glutamine and cancer: cell biology, physiology,
58. Liśkiewicz, A. D. et al. Long-​term high fat ketogenic Lipid Mediat. 127, 9–13 (2016). and clinical opportunities. J. Clin. Invest. 123,
diet promotes renal tumor growth in a rat model 85. Sansbury, B. E., Bhatnagar, A. & Hill, B. G. Impact 3678–3684 (2013).
of tuberous sclerosis. Sci. Rep. 6, 21807 (2016). of nutrient excess and endothelial nitric oxide 109. Sellers, K. et al. Pyruvate carboxylase is critical for
59. Xia, S. et al. Prevention of dietary-​fat-fueled synthase on the plasma metabolite profile in mice. non-​small-cell lung cancer proliferation. J. Clin. Invest.
ketogenesis attenuates BRAF V600E tumor growth. Front. Physiol. 5, 453 (2014). 125, 687–698 (2015).
Cell Metab. 25, 358–373 (2017). 86. Lai, Y. S. et al. Mass-​spectrometry-based serum 110. Tardito, S. et al. Glutamine synthetase activity fuels
60. Kang, H. B. et al. Metabolic rewiring by oncogenic metabolomics of a c57bl/6j mouse model of high-​ nucleotide biosynthesis and supports growth of
BRAF V600E links ketogenesis pathway to BRAF-​MEK1 fat-diet-​induced non-​alcoholic fatty liver disease glutamine-​restricted glioblastoma. Nat. Cell Biol. 17,
signaling. Mol. Cell 59, 345–358 (2015). development. J. Agric Food Chem. 63, 7873–7884 1556–1568 (2015).
61. Morscher, R. J. et al. Inhibition of neuroblastoma (2015). 111. Raffaghello, L. et al. Starvation-​dependent differential
tumor growth by ketogenic diet and/or calorie 87. Stone, K. P. et al. Compromised responses to dietary stress resistance protects normal but not cancer cells
restriction in a CD1-nu mouse model. PLOS ONE 10, methionine restriction in adipose tissue but not liver against high-​dose chemotherapy. Proc. Natl Acad. Sci.
e0129802 (2015). of ob/ob mice. Obesity (Silver Spring) 23, 1836–1844 USA 105, 8215–8220 (2008).
62. Woolf, E. C., Syed, N. & Scheck, A. C. Tumor metabolism, (2015). 112. Schroll, M. M., Liu, X., Herzog, S. K., Skube, S. B.
the ketogenic diet and β-​hydroxybutyrate: novel 88. Kokkinakis, D. M., Schold, S. C. J., Hori, H. & & Hummon, A. B. Nutrient restriction of glucose
approaches to adjuvant brain tumor therapy. Front. Nobori, T. Effect of long-​term depletion of plasma or serum results in similar proteomic expression
Mol. Neurosci. 9, 122 (2016). methionine on the growth and survival of human brain changes in 3D colon cancer cell cultures. Nutr. Res.
63. Zhou, W. et al. The calorically restricted ketogenic tumor xenografts in athymic mice. Nutr. Cancer 29, 36, 1068–1080 (2016).
diet, an effective alternative therapy for malignant 195–204 (1997). 113. Lashinger, L. M. et al. Starving cancer from the
brain cancer. Nutr. Metab. (Lond) 4, 5 (2007). 89. Lees, E. K. et al. Direct comparison of methionine outside and inside: separate and combined effects
64. Seyfried, T. N., Sanderson, T. M., El-​Abbadi, M. M., restriction with leucine restriction on the metabolic of calorie restriction and autophagy inhibition on
McGowan, R. & Mukherjee, P. Role of glucose and health of C57BL/6J mice. Sci. Rep. 7, 9977 (2017). Ras-​driven tumors. Cancer Metab. 4, 18 (2016).
ketone bodies in the metabolic control of experimental 90. Elshorbagy, A. K. et al. Cysteine supplementation 114. Bianchi, G. et al. Fasting induces anti-​Warburg effect
brain cancer. Br. J. Cancer. 89, 1375–1382 (2003). reverses methionine restriction effects on rat adiposity: that increases respiration but reduces atp-​synthesis
65. Maurer, G. D. et al. Differential utilization of ketone significance of stearoyl-​coenzyme A desaturase. to promote apoptosis in colon cancer models.
bodies by neurons and glioma cell lines: a rationale J. Lipid Res. 52, 104–112 (2011). Oncotarget 6, 11806–11819 (2015).
for ketogenic diet as experimental glioma therapy. 91. Huang, T. H. et al. A methionine-​restricted diet and 115. Zhang, J. et al. Low ketolytic enzyme levels in tumors
BMC Cancer 11, 315 (2011). endurance exercise decrease bone mass and extrinsic predict ketogenic diet responses in cancer cell lines
66. Shukla, S. K. et al. Metabolic reprogramming induced strength but increase intrinsic strength in growing in vitro and in vivo. J. Lipid Res. 59, 625–634 (2018).
by ketone bodies diminishes pancreatic cancer male rats. J. Nutr. 144, 621–630 (2014). 116. Huang, D. et al. Hepatocellular carcinoma redirects to
cachexia. Cancer Metab. 2, 18 (2014). 92. Fukumura, D., Duda, D. G., Munn, L. L. & Jain, R. K. ketolysis for progression under nutrition deprivation
67. Wheatley, K. E. et al. Low-​carbohydrate diet versus Tumor microvasculature and microenvironment: stress. Cell Res. 26, 1112–1130 (2016).
caloric restriction: effects on weight loss, hormones, novel insights through intravital imaging in pre-​clinical 117. Buescher, J. M. et al. A roadmap for interpreting 13C
and colon tumor growth in obese mice. Nutr. Cancer models. Microcirculation 17, 206–225 (2010). metabolite labeling patterns from cells. Curr. Opin.
60, 61–68 (2008). 93. Pan, M. et al. Regional glutamine deficiency in Biotechnol. 34, 189–201 (2015).
68. Allen, B. G. et al. Ketogenic diets enhance oxidative tumours promotes dedifferentiation through 118. Chong, J. et al. MetaboAnalyst 4.0: towards more
stress and radio-​chemo-therapy responses in lung inhibition of histone demethylation. Nat. Cell Biol. transparent and integrative metabolomics analysis.
cancer xenografts. Clin. Cancer Res. 19, 3905–3913 18, 1090–1101 (2016). Nucleic Acids Res. 46, W486–W494 (2018).
(2013). 94. Wiig, H. & Swartz, M. A. Interstitial fluid and lymph 119. Sergushichev, A. A. et al. GAM: a web-​service for
69. Pascual, G. et al. Targeting metastasis-​initiating cells formation and transport: physiological regulation and integrated transcriptional and metabolic network
through the fatty acid receptor CD36. Nature 541, roles in inflammation and cancer. Physiol. Rev. 92, analysis. Nucleic Acids Res. 44, W194–W200 (2016).
41–45 (2017). 1005–1060 (2012). 120. Hui, S. et al. Glucose feeds the TCA cycle via circulating
70. Chen, M. et al. An aberrant SREBP-​dependent 95. Wiig, H., Aukland, K. & Tenstad, O. Isolation of lactate. Nature 551, 115–118 (2017).
lipogenic program promotes metastatic prostate interstitial fluid from rat mammary tumors by a 121. Valkenburg, K. C., de Groot, A. E. & Pienta, K. J.
cancer. Nat. Genet. 50, 206–218 (2018). centrifugation method. Am. J. Physiol. Heart Circ. Targeting the tumour stroma to improve cancer
71. Knott, S. R. V. et al. Asparagine bioavailability governs Physiol. 284, H416–H424 (2003). therapy. Nat. Rev. Clin. Oncol. 15, 366–381 (2018).
metastasis in a model of breast cancer. Nature 554, 96. Haslene-​Hox, H. et al. A new method for isolation 122. Lyssiotis, C. A. & Kimmelman, A. C. Metabolic
378–381 (2018). of interstitial fluid from human solid tumors applied interactions in the tumor microenvironment.
72. Patti, G. J., Yanes, O. & Siuzdak, G. Innovation: to proteomic analysis of ovarian carcinoma tissue. Trends Cell Biol. 27, 863–875 (2017).
metabolomics: the apogee of the omics trilogy. PLOS ONE 6, e19217 (2011). 123. Ying, M., Guo, C. & Hu, X. The quantitative
Nat. Rev. Mol. Cell Biol. 13, 263–269 (2012). 97. Ho, P. C. et al. Phosphoenolpyruvate is a metabolic relationship between isotopic and net contributions
73. Collet, T. H. et al. A metabolomic signature of acute checkpoint of anti-​tumor T Cell Res.ponses. Cell 162, of lactate and glucose to the TCA cycle. J. Biol. Chem.
caloric restriction. J. Clin. Endocrinol. Metab. 102, 1217–1228 (2015). 294, 9615–9630 (2019).
4486–4495 (2017). 98. Burgess, E. A. & Sylven, B. Glucose, lactate, and lactic 124. Timmers, S. et al. Calorie restriction-​like effects
74. Margolis, L. M. et al. Calorie restricted high protein dehydrogenase activity in normal interstitial fluid and of 30 days of resveratrol supplementation on energy
diets downregulate lipogenesis and lower intrahepatic that of solid mouse tumors. Cancer Res. 22, 581–588 metabolism and metabolic profile in obese humans.
triglyceride concentrations in male rats. Nutrients 8, (1962). Cell Metab. 14, 612–622 (2011).
E571 (2016). 99. Gullino, P. M., Clark, S. H. & Grantham, F. H. 125. Pernicova, I. & Korbonits, M. Metformin–mode of
75. Miller, K. N. et al. Aging and caloric restriction impact The interstitial fluid of solid tumors. Cancer Res. 24, action and clinical implications for diabetes and cancer.
adipose tissue, adiponectin, and circulating lipids. 780–794 (1964). Nat. Rev. Endocrinol. 10, 143–156 (2014).
Aging Cell 16, 497–507 (2017). 100. Hensley, C. T. et al. Metabolic heterogeneity in human 126. Fang, Y. et al. Duration of rapamycin treatment has
76. Selman, C. et al. Coordinated multitissue transcriptional lung tumors. Cell 164, 681–694 (2016). differential effects on metabolism in mice. Cell Metab.
and plasma metabonomic profiles following acute 101. Poff, A. M., Ari, C., Arnold, P., Seyfried, T. N. & 17, 456–462 (2013).
caloric restriction in mice. Physiol. Genomics 27, D’Agostino, D. P. Ketone supplementation decreases 127. Ohe, K., Morris, H. P. & Weinhouse, S. Beta-​
187–200 (2006). tumor cell viability and prolongs survival of mice with hydroxybutyrate dehydrogenase activity in liver
77. Raeini-​Sarjaz, M., Vanstone, C. A., Papamandjaris, A. A., metastatic cancer. Int. J. Cancer 135, 1711–1720 and liver tumors. Cancer Res. 27, 1360–1371
Wykes, L. J. & Jones, P. J. Comparison of the effect (2014). (1967).
of dietary fat restriction with that of energy restriction 102. Birsoy, K. et al. Metabolic determinants of cancer 128. Tisdale, M. J. & Brennan, R. A. Loss of acetoacetate
on human lipid metabolism. Am. J. Clin. Nutr. 73, cell sensitivity to glucose limitation and biguanides. coenzyme A transferase activity in tumours of
262–267 (2001). Nature 508, 108–112 (2014). peripheral tissues. Br. J. Cancer. 47, 293–297
78. Appel, L. J. et al. Effects of protein, monounsaturated 103. Schwarz, J. M., Linfoot, P., Dare, D. & Aghajanian, K. (1983).
fat, and carbohydrate intake on blood pressure and Hepatic de novo lipogenesis in normoinsulinemic 129. Patel, M. S., Russell, J. J. & Gershman, H.
serum lipids: results of the OmniHeart randomized and hyperinsulinemic subjects consuming high-​fat, Ketone-​body metabolism in glioma and neuroblastoma
trial. JAMA 294, 2455–2464 (2005). low-​carbohydrate and low-​fat, high-​carbohydrate cells. Proc. Natl Acad. Sci. USA 78, 7214–7218
79. Ma, Y. et al. Association between carbohydrate intake isoenergetic diets. Am. J. Clin. Nutr. 77, 43–50 (1981).
and serum lipids. J. Am. Coll. Nutr. 25, 155–163 (2003). 130. Magee, B. A., Potezny, N., Rofe, A. M. & Conyers, R. A.
(2006). 104. Ikemoto, S. et al. High-​fat diet-​induced hyperglycemia The inhibition of malignant cell growth by ketone
80. Kennedy, A. R. et al. A high-​fat, ketogenic diet induces and obesity in mice: differential effects of dietary oils. bodies. Aust. J. Exp. Biol. Med. Sci. 57, 529–539
a unique metabolic state in mice. Am. J. Physiol. Metabolism 45, 1539–1546 (1996). (1979).
Endocrinol. Metab. 292, E1724–E1739 (2007). 105. Jang, C. et al. The small intestine converts dietary 131. Rodrigues, L. M. et al. The action of β-​hydroxybutyrate
81. Douris, N. et al. Adaptive changes in amino acid fructose into glucose and organic acids. Cell Metab. on the growth, metabolism and global histone H3
metabolism permit normal longevity in mice consuming 27, 351–361.e3 (2018). acetylation of spontaneous mouse mammary
a low-​carbohydrate ketogenic diet. Biochim. Biophys. 106. Balakumar, M. et al. High-​fructose diet is as detrimental tumours: evidence of a β-​hydroxybutyrate paradox.
Acta. 1852, 2056–2065 (2015). as high-​fat diet in the induction of insulin resistance and Cancer Metab. 5, 4 (2017).

www.nature.com/nrc
Perspectives

132. Xu, X. et al. Overview of the development of in melanoma and glioma. AntiCancer Res. 29, desaturase as a target for cancer therapy. Mol. Cancer
glutaminase inhibitors: achievements and future 5235–5240 (2009). Res. 9, 1551–1561 (2011).
directions. J. Med. Chem. 62, 1096–1115 (2019). 146. Durando, X. et al. Dietary methionine restriction with 158. Peck, B. et al. Inhibition of fatty acid desaturation is
133. Lien, E. C., Lyssiotis, C. A. & Cantley, L. C. FOLFOX regimen as first line therapy of metastatic detrimental to cancer cell survival in metabolically
Metabolic reprogramming by the PI3K-​Akt-mTOR colorectal cancer: a feasibility study. Oncol. 78, compromised environments. Cancer Metab. 4, 6
pathway in cancer. Recent Results Cancer Res. 207, 205–209 (2010). (2016).
39–72 (2016). 147. Di Biase, S. et al. Fasting regulates EGR1 and protects 159. Peck, B. & Schulze, A. Lipid desaturation - the next
134. Kanarek, N. et al. Histidine catabolism is a major from glucose- and dexamethasone-​dependent step in targeting lipogenesis in cancer. FEBS J. 283,
determinant of methotrexate sensitivity. Nature 559, sensitization to chemotherapy. PLOS Biol. 15, 2767–2778 (2016).
632–636 (2018). e2001951 (2017). 160. Reeves, P. G., Nielsen, F. H. & Fahey, G. C. AIN-93
135. Huennekens, F. M. The methotrexate story: 148. Obrist, F. et al. Metabolic vulnerability of cisplatin-​ purified diets for laboratory rodents: final report of
a paradigm for development of cancer resistant cancers. EMBO J. 37, e98597 (2018). the American Institute of Nutrition ad hoc writing
chemotherapeutic agents. Adv. Enzyme Regul. 34, 149. Gravel, S. P. et al. Serine deprivation enhances committee on the reformulation of the AIN-76A
397–419 (1994). antineoplastic activity of biguanides. Cancer Res. 74, rodent diet. J. Nutr. 123, 1939–1951 (1993).
136. Nencioni, A., Caffa, I., Cortellino, S. & Longo, V. D. 7521–7533 (2014). 161. Ellacott, K. L., Morton, G. J., Woods, S. C., Tso, P. &
Fasting and cancer: molecular mechanisms and 150. Ricci, M. R. & Ulman, E. A. Laboratory animal diets: Schwartz, M. W. Assessment of feeding behavior in
clinical application. Nat. Rev. Cancer 18, 707–719 a critical part of your in vivo research. Research Diets laboratory mice. Cell Metab. 12, 10–17 (2010).
(2018). https://researchdiets.com/system/resources/W1si 162. Pugh, T. D., Klopp, R. G. & Weindruch, R. Controlling
137. Hoshiya, Y., Kubota, T., Matsuzaki, S. W., Kitajima, M. ZiIsIjIwMTgvMDMvMjkvNzF0NjQ5OXcwZl9 caloric consumption: protocols for rodents and rhesus
& Hoffman, R. M. Methionine starvation modulates MYWJvcmF0b3J5X0FuaW1hbF9EaWV0c19f monkeys. Neurobiol. Aging 20, 157–165 (1999).
the efficacy of cisplatin on human breast cancer QV9Dcml0aWNhbF9QYXJ0X29mX3lvdXJfS
in nude mice. AntiCancer Res. 16, 3515–3517 W5fdml2b19SZXNlYXJjaF93ZWIucGRmIl1d/ Acknowledgements
(1996). Laboratory_Animal_Diets_%20A_Critical_Part_ The authors thank members of the Vander Heiden laboratory
138. Poirson-​Bichat, F., Gonçalves, R. A., Miccoli, L., of_your_In_vivo_Research_web.pdf (2005). for thoughtful discussions and comments on the manuscript.
Dutrillaux, B. & Poupon, M. F. Methionine depletion 151. Chen, Y. et al. The influence of dietary lipid composition E.C.L. is a Damon Runyon Fellow supported by the Damon
enhances the antitumoral efficacy of cytotoxic on liver mitochondria from mice following 1 month of Runyon Cancer Research Foundation (DRG-2299-17). M.G.V.H.
agents in drug-​resistant human tumor xenografts. calorie restriction. BioSci. Rep. 33, 83–95 (2012). is supported by the Emerald Foundation, the Lustgarten
Clin. Cancer Res. 6, 643–653 (2000). 152. Abbott, S. K., Else, P. L., Atkins, T. A. & Hulbert, A. J. Foundation, SU2C, the Ludwig Center at MIT, the US National
139. Lee, C. et al. Fasting cycles retard growth of tumors Fatty acid composition of membrane bilayers: Cancer Institute, the MIT Center for Precision Cancer Medicine
and sensitize a range of cancer cell types to importance of diet polyunsaturated fat balance. and a Faculty Scholars award from the Howard Hughes Medical
chemotherapy. Sci. Transl. Med. 4, 124ra27 (2012). Biochim. Biophys. Acta. 1818, 1309–1317 (2012). Institute.
140. de Groot, S. et al. The effects of short-​term fasting 153. Tosi, F., Sartori, F., Guarini, P., Olivieri, O. &
on tolerance to (neo) adjuvant chemotherapy in Martinelli, N. Delta-5 and delta-6 desaturases: Author contributions
HER2-negative breast cancer patients: a randomized crucial enzymes in polyunsaturated fatty acid-​related Both authors contributed to the discussion of manuscript
pilot study. BMC Cancer 15, 652 (2015). pathways with pleiotropic influences in health and content, writing of the manuscript and reviewing or editing of
141. Dorff, T. B. et al. Safety and feasibility of fasting in disease. Adv. Exp. Med. Biol. 824, 61–81 (2014). the manuscript before submission.
combination with platinum-​based chemotherapy. 154. Ackerman, D. et al. triglycerides promote lipid
BMC Cancer 16, 360 (2016). homeostasis during hypoxic stress by balancing Competing interests
142. Safdie, F. M. et al. Fasting and cancer treatment in fatty acid saturation. Cell Rep. 24, 2596–2605.e5 M.G.V.H. is a consultant for and scientific advisory board mem-
humans: a case series report. Aging (Albany NY) 1, (2018). ber of Agios Pharmaceuticals, Aeglea Biotherapeutics and
988–1007 (2009). 155. Kamphorst, J. J. et al. Hypoxic and Ras-​transformed Auron Therapeutics. E.C.L. reports no competing interests.
143. Bauersfeld, S. P. et al. The effects of short-​term fasting cells support growth by scavenging unsaturated fatty
on quality of life and tolerance to chemotherapy in acids from lysophospholipids. Proc. Natl Acad. Sci. Peer review information
patients with breast and ovarian cancer: a randomized USA 110, 8882–8887 (2013). Nature Reviews Cancer thanks L. Hodson, V. Longo and the
cross-​over pilot study. BMC Cancer 18, 476 (2018). 156. Serini, S., Piccioni, E., Merendino, N. & Calviello, G. other, anonymous, reviewer(s) for their contribution to the peer
144. Hoffman, R. M. Clinical studies of methionine-​restricted Dietary polyunsaturated fatty acids as inducers of review of this work.
diets for cancer patients. Methods Mol. Biol. 1866, apoptosis: implications for cancer. Apoptosis 14,
95–105 (2019). 135–152 (2009). Publisher’s note
145. Thivat, E. et al. Phase II trial of the association 157. Roongta, U. V. et al. Cancer cell dependence on Springer Nature remains neutral with regard to jurisdictional
of a methionine-​free diet with cystemustine therapy unsaturated fatty acids implicates stearoyl-​CoA claims in published maps and institutional affiliations.

Nature Reviews | Cancer

You might also like