You are on page 1of 15

REVIEWS

M e ta b o l i c s i g n a l l i n g

Metabolic regulation of cell growth


and proliferation
Jiajun Zhu    and Craig B. Thompson   *
Abstract | Cellular metabolism is at the foundation of all biological activities. The catabolic
processes that support cellular bioenergetics and survival have been well studied. By contrast,
how cells alter their metabolism to support anabolic biomass accumulation is less well
understood. During the commitment to cell proliferation, extensive metabolic rewiring must
occur in order for cells to acquire sufficient nutrients such as glucose, amino acids, lipids and
nucleotides, which are necessary to support cell growth and to deal with the redox challenges
that arise from the increased metabolic activity associated with anabolic processes. Defining the
mechanisms of this metabolic adaptation for cell growth and proliferation is now a major focus of
research. Understanding the principles that guide anabolic metabolism may ultimately enhance
ways to treat diseases that involve deregulated cell growth and proliferation, such as cancer.

Non-essential amino acids


Cell proliferation requires the accumulation of intra­ cell proliferation and then consider how cells maintain
(NEAAs). Amino acids that can cellular biomass, such as proteins and lipids, in order growth in nutrient-scarce environments such as wound
be synthesized endogenously to produce daughter cells. At the same time, DNA repli­ beds and in poorly vascularized tumour regions.
and often participate in cation must occur to pass along the genetic informa­
processes beyond protein
tion. Accordingly, net production of proteins, lipids and Regulation of glucose uptake
translation, such as nucleotide
synthesis. nucleic acids is essential for a successful replicative cell In addition to the use of glucose as an energy source
division. The biosynthesis of these macromolecules is for ATP generation, proliferating cells often display
Anaplerosis achieved mainly through a network of cellular metabolic enhanced uptake of glucose, providing an important
The set of biochemical
pathways that direct the acquisition and utilization of source of carbon to support lipid production and the
reactions that replenish the
intermediates of metabolic
various sources of nutrients. Characterizing the princi­ biosynthesis of nucleotides and non-essential amino acids
pathways. ples underlying these cellular biosynthetic pathways can (NEAAs), which occurs via redirection of metabolites of
thus provide important insights into the understanding glycolysis and the tricarboxylic acid (TCA) cycle (FiG. 1;
of cell growth and proliferation. Supplementary Box 1). To replenish TCA cycle interme­
Unlike unicellular organisms, which directly sense and diates that are used for the production of biomass, cells
scavenge nutrients from the environment1–3, metazoan cells use anaplerosis, and an important anaplerotic substrate
do not generally take up nutrients in a cell-autonomous is glutamine4. Characterization of how metazoan cells
manner. In animal cells, nutrient acquisition is directed regulate glucose uptake and its utilization is important to
primarily by growth factor signalling (Box  1) . The the understanding of how cellular metabolism is rewired
directed uptake of nutrients then stimulates intracellular to support anabolic growth.
nutrient-sensing kinases, establishing signalling cascades
that redirect nutrients from catabolic pathways (aimed at Signalling cues instruct glucose uptake. In animal
molecule breakdown for energy production) to anabolic cells, glucose uptake is triggered non-cell-­autonomously.
pathways (aimed at biosynthesis of cellular components). Metazoan cell growth is primarily stimulated by growth
Recent studies have shed new light on how cells repro­ factor signalling (Box  1), and cells can only acquire
gramme their metabolism from ­catabolism to anabolism large quantities of glucose as a result of the activation
Department of Cancer to fuel cell proliferation. of these signalling pathways. Various growth factors,
Biology and Genetics, In this Review, we examine the regulation of nutrient such as insulin, vascular endothelial growth factor
Memorial Sloan Kettering uptake and utilization when cells are instructed to pro­ (VEGF) and epidermal growth factor (EGF), serve as
Cancer Center, New York,
NY, USA.
liferate by physiological cues, such as in the context of the primary signalling cues that direct glucose uptake. In
tissue regeneration, and when cells acquire oncogenic insulin-responsive cells, such as those in the muscle, liver
*e-mail: thompsonc@
mskcc.org mutations that stimulate cell proliferation. We highlight and fat tissues, most of this glucose is converted for stor­
https://doi.org/10.1038/ the regulation of glucose uptake, amino acid acquisition age into glycogen and fats (primarily triglycerides stored
s41580-019-0123-5 and lipid and nucleotide synthesis during homeostatic in the adipose tissue). However, in most other cells, an

Nature Reviews | Molecular Cell Biology


Reviews

Minimal medium increase in glucose metabolism following growth factor acid, nucleotide and fatty acid biosynthesis12 (Box 1;
Medium that contains the signalling leads to accumulation of biomass5–8. Glucose Supplementary Box 1). Upon withdrawal of growth
minimum nutrients possible for uptake is often increased in growth-factor-stimulated factor, T cells rapidly undergo apoptosis as a result of
the growth of cells. cells. This is largely achieved through a series of kinase- reduction in mitochondrial membrane potential and
mediated signalling events, including the activation of cellular ATP level. However, cell survival can be rescued
receptor tyrosine kinases (RTKs) and the downstream by ectopic expression of GLUT1 and hexo­kinase14. These
PI3K and AKT (also known as protein kinase B (PKB)) studies demonstrate that glucose uptake and utilization
signalling cascade (Box 1). are direct results of extracellular stimuli and that main­
Studies of T cell activation have provided good exam­ taining glucose influx and metabolism is essential to
ples of how PI3K and AKT signalling informs a cell to sustain cell survival and growth.
take up and utilize glucose in support of cell growth. In
the context of T cells, activation of T cell receptor (TCR) Utilizing glucose for cell growth. Stimulation of rest­
upon antigen stimulation leads to the tyrosine phosphory­ ing T cells by TCR and CD28 induces a drastic increase
lation of the cytoplasmic tail region of co-stimulatory in glucose uptake, and this uptake supports the clonal
receptor CD28, which — analogous to RTKs — is able expansion of antigen-stimulated T cells15. Augmented
to initiate a number of signalling cascades including the glucose influx leads to a high glycolytic rate. However,
PI3K–AKT axis9,10. The activation of AKT promotes only a small fraction of glucose is fully oxidized in the
expression and plasma membrane localization of the TCA cycle (Supplementary Box 1) and serves as a source
glucose transporter 1 (GLUT1)11–13. In addition, AKT for ATP production via oxidative phosphorylation. In
increases the cellular activity of the glycolytic enzyme fact, the majority of carbon in glucose is converted into
hexokinase such that the imported glucose is captured by lactate and secreted. This has long been considered para­
glycolysis as well as by other pathways to support amino doxical. For the past 100 years, cellular nutrient uptake
has been considered a demand-driven system based
on biochemical studies performed primarily in bacte­
Box 1 | growth factor signalling ria grown in minimal medium. Under such conditions,
Growth factors are signalling molecules that promote cell growth, proliferation or glycolysis is regulated by a declining level of ATP, and
differentiation. typical examples of growth factors include insulin, epidermal growth oxidative phosphorylation is stimulated primarily by a
factor (eGF), fibroblast growth factor (FGF), erythropoietin (ePO), platelet-derived rise in ADP. However, the glucose uptake in metazoans
growth factor (PDGF), transforming growth factors (tGFs) and cytokines. Growth factors is a supply-driven system in which the cellular glucose
are usually secreted by specific organs and cell types to promote particular physiological uptake is primarily stimulated by growth factor signal­
processes. For example, insulin is produced by the β-pancreatic islet cells when the ling. When growth factor-stimulated cells take up more
body nutrient levels are high, and it stimulates glucose uptake by liver, fat and skeletal glucose than they require, excess glycolytic intermedi­
muscle cells where glucose can be converted into glycogen or triglycerides for energy
ates are diverted into pathways that support the produc­
storage8. upon tissue injury, eGF, FGF and a number of other growth factors are secreted
tion of the NEAAs, nucleotides and lipids required for
by local immune cells and fibroblasts to promote tissue regeneration that involves the
proliferation of keratinocytes and endothelial cells6. cell growth. Any excess glycolytic end products are dealt
Growth-factor-stimulated cells often display increased ability to take up nutrients. with by converting pyruvate into lactate and secreting
receptor tyrosine kinase (rtK) signalling is a typical downstream mediator of growth lactate back into the extracellular environment (FiG. 1).
factor stimulation. Most growth factors that regulate metabolism bind to cognate rtKs, The metabolic phenomenon by which rapidly pro­
and this binding results in the autophosphorylation of the receptor to initiate a series liferating cells in a nutrient-rich environment continue
of kinase-mediated signalling events. Members of the Pi3K family are major targets of to take up and metabolize glucose in excess of their ana­
rtKs. upon interaction with the autophosphorylated form of rtK, Pi3K is activated bolic requirements is known as aerobic glycolysis (also
and converts the plasma membrane lipid phosphatidylinositol-4,5-bisphosphate known as the Warburg effect). In fact, this phenom­
(Ptdins(4,5)P2) into phosphatidylinositol-3,4,5-trisphosphate (Ptdins(3,4,5)P3).
enon has been known for almost a century, with initial
Local enrichment of Ptdins(3,4,5)P3 serves as the subsequent signalling cue to bring
observations suggesting that tumour cells display a much
together kinases including phosphoinositide-dependent kinase 1 (PDK1) and aKt155.
PDK1 can phosphorylate the regulatory loop of aKt and promote aKt activation. higher rate of glucose consumption than their normal
aKt is a serine/threonine kinase, for which a variety of downstream targets have been counterparts and that, in tumour cells, glucose is mainly
identified, including glucose uptake through glucose transporter 1 (GLut1) and utilized via glycolysis even in the presence of adequate
induction of glycolysis by activation of hexokinase (HK), which converts glucose into oxygen16,17. Unlike normal cells that are instructed to
glucose 6-phosphate (G6P) (see the figure). together, activation of aKt targets supports proliferate by extracellular signals, most cancer cells have
an anabolic growth phenotype156,157. this process is under tight control, as Ptdins(3,4,5)P3 acquired the ability to take up glucose cell-autonomously
can be dephosphorylated and converted back into Ptdins(4,5)P2 through the lipid through the activation of oncogenes (FiG. 1) and hence
phosphatase activity of PteN158. this negative regulatory mechanism, among others, acquire much more glucose than they require for their
dampens the Pi3K–aKt pathway activity and ensures that normal cells can commit
oxidative metabolism. Cancer genome sequencing has
themselves to proliferation only when persistent growth factor signalling is present.
revealed that a large portion of human cancers harbour
Glucose genetic alterations in the genes encoding members of
Growth
factor the RTK family that increase their kinase activities18.
RTK GLUT1 For example, EGF receptor mutation or amplification
occurs in 50% of patients with non-small-cell lung can­
PtdIns(4,5)P2 PtdIns(3,4,5)P3 PDK1 AKT cer (NSCLC) in Asia and 15% of patients with NSCLC
P P PTEN P in Western countries19. Human epidermal growth factor
HK receptor 2 (HER2; also known as ERBB2) is amplified in
PI3K G6P up to 20% of patients with breast cancer20. In addition

www.nature.com/nrm
Reviews

Growth factors Oncogenes Quantitative metabolic flux analyses at the organismal


(normal cells) Uptake stimulation (cancer cells; growth
Glucose
factor-independent) level have led to new insights into glucose metabolism
GLUT1 in tumours in vivo27,28. It has been shown that lactate
produced by tumour cells via aerobic glycolysis can be
consumed by nonproliferating tumour cells and the
Protein surrounding stromal cells in the tumour microenviron­
Glucose
synthesis ment, which direct lactate to the TCA cycle, using it as
Pentose Promoted Serine
a primary source of carbon. Similar observations have
phosphate in cancer biosynthesis also been made in normal tissues, and it has been sug­
pathway pathway gested that, in some tissues, the input of glucose into the
Ribose Serine
TCA cycle is mostly indirect — via circulating lactate
Promoted produced by other cells28.
in cancer
Pyruvate MCT
Lactate Acquisition of amino acids
Circulating
lactate In addition to the uptake of glucose as a carbon source,
Aspartate proliferating cells also need a substantial amount of
Uptake by
neighbouring amino acids in order to increase protein content, which
cells to fuel makes up more than half of a cell’s dry mass29. Through
Citrate TCA cycle
Nucleotide TCA a network of nutrient-responsive pathways, cells main­
synthesis cycle
tain a battery of amino acids to charge tRNAs for protein
Fatty acid Protein translation, to support the biosynthesis of macromol­
synthesis synthesis ecules including nucleotides and to fuel bioenergetics
Mitochondrion
such as through the catabolism of certain amino acids
Anaplerosis to produce acetyl-CoA. In proliferating cells, metabolic
Glutamine
programmes are often rewired to actively acquire amino
Glutamine
acids from the extracellular space to sustain biomass
Proliferating cell accumulation. In addition, opportunistic pathways can
be utilized to scavenge protein when free amino acids are
Fig. 1 | glucose uptake and utilization. In order to support cell growth and
proliferation, glucose uptake is promoted by the growth-factor-signalling pathways limiting in the extracellular environment.
in normal cells or by oncogenic mutations in cancer cells, which may render growth
signalling pathways constantly active.  Glucose is an important carbon source in Sensing and importing amino acids. Similar to glu­
energy-generating pathways of oxidative metabolism, fuelling the tricarboxylic acid cose, growth factor signalling can also directly promote
(TCA) cycle and subsequent oxidative phosphorylation in mitochondria (not shown). amino acid uptake and utilization (FiG. 2). The mecha­
In proliferating cells, intermediates of glucose metabolism are often diverted from nistic target of rapamycin complex 1 (mTORC1) is a
glycolysis and the TCA cycle (dashed arrows) and used for biosynthetic purposes such as central coordinator of amino acid availability and allo­
the production of nucleotides, amino acids and fatty acids. The TCA cycle is kept active cation30. Increased AKT activity downstream of growth
by anaplerotic reactions involving glutamine, which — beyond supporting anabolic factor signalling results in phosphorylation of tuberous
reactions — replenishes TCA cycle intermediates that are diverted from the cycle to
sclerosis complex, subunit 2 (TSC2), and subsequently
support anabolism. As a result of aerobic glycolysis, a large portion of glucose carbon is
also converted into lactate and secreted. Cancer-associated adaptations are highlighted dislocation of the TSC complex from the lysosome31–33.
in red. GLUT1, glucose transporter 1; MCT, monocarboxylate transporter. Dissociation of TSC from its lysosome-resident target,
RAS homologue enriched in brain (RHEB), leads to the
derepression of RHEB activity and results in the acti­
to RTKs, the PIK3CA gene, encoding the catalytic sub­ vation of mTORC1 kinase activity34 (FiG. 2). A primary
unit of PI3K, is one of the most frequently mutated functional output of mTORC1 activation is the increase
genes in cancer, particularly in breast, colon and liver in protein synthesis, which is achieved largely through
cancers21,22. Loss-of-function mutations in the PTEN direct phosphorylation of p70S6 kinase 1 (S6K1) and
Positron-emission-
tomography-based imaging gene — a negative regulator of PI3K signalling — are eIF4E binding protein 1 (4EBP1) by mTORC1 (ref.30). To
An imaging technique used also highly prevalent in prostate and endometrial can­ complement the demand for increased protein transla­
clinically to observe metabolic cers23. These genetic events are selected, at least in part, tion, concerted AKT and mTORC1 activation induces
processes for disease diagnosis to allow tumour cells to constantly acquire glucose and and sustains cell surface expression of amino acid trans­
or monitoring.
other nutrients from the environment, independently of porters35. In addition, through S6K1 kinase activity,
p70S6 kinase 1 growth factor stimulation. mTORC1 can induce ribosome biogenesis in order to
(S6K1). A kinase downstream Deregulated glucose uptake has emerged as a hall­ sustain the expanded protein translation capacity36.
of the mTOR complex 1 mark of cancer metabolism24. In fact, positron-emission- Besides growth-factor-signalling inputs, mTORC1
signalling pathway, the
tomography-based imaging of 18F-fluorodeoxyglucose activity is tightly controlled by the availability of amino
activation of which can lead to
increased protein synthesis. uptake (FDG–PET) is widely used in the clinic for acids. When sufficient amounts of amino acids are
tumour monitoring25. When studied in tissue culture, present in the cells, the heterodimeric RAG GTPases
eIF4E binding protein 1 cancer cells utilize glucose to generate glycolytic and promote the localization of mTORC1 to the lysosome,
(4EBP1). A protein translation TCA cycle intermediates in order to support biomolecu­ where it is in proximity to RHEB37. Regulation of the
repressor. Its activity is
inhibited by phosphorylation
lar synthesis. Despite this, most cultured cancer cell lines RAG GTPases and hence mTORC1 activity by amino
mediated by mTOR complex 1 secrete the majority of the glucose they take up as lac­ acid levels is achieved through specific amino acid sen­
to increase protein synthesis. tate, which accumulates in the tissue culture medium26. sor proteins. For example, Sestrin-2 (encoded by SESN2)

Nature Reviews | Molecular Cell Biology


Reviews

Integrated stress response


was identified as a leucine sensor38,39. When the cellular the GATOR2–GATOR1 axis. Overall, the intracellu­
A mechanism that usually leucine level decreases, Sestrin-2 can bind and inhibit lar amino acid sensors directly influence amino acid
suppresses general protein GATOR2, leading to the activation of the GATOR1 consumption by decreasing mTORC1 stimulation of
synthesis but promotes the complex, which represses the RAG GTPases and hence ­translation when these amino acids become limiting40,41.
expression of specific
transcription factors to mediate
precludes full mTORC1 activation37 (FiG. 2). In addition, In addition to the suppression of mTORC1 activ­
cellular stress response. CASTOR1 was identified as a sensor of cytosolic argi­ ity, a reduction in amino acid levels also provokes the
nine40, and SAMTOR (encoded by BMT2) was shown to integrated stress response. As a considerable amount
be a sensor of S-adenosyl methionine and hence indic­ of tRNA becomes uncharged upon amino acid defi­
ative of the cellular methionine level41 (FiG. 2). Similarly ciency, general control nonderepressible 2 (GCN2)
to Sestrin-2, both CASTOR1 and SAMTOR function kinase is activated, which phosphorylates eukary­
as negative regulators of the RAG GTPases through otic initiation factor 2α (eIF2α) to attenuate global

Integrated
stress response
Leucine
Sestrin-2 GCN2 Amino acid
deficiency
GATOR2 tRNA ATF4
Arginine synthetases
CASTOR1
Adaptation and
recovery from stress Glutamate
ASNS PHGDH
Methionine PSAT1
GATOR1 Protein x(c)(–)
translation
SAM SAMTOR Asparagine Cystine
synthesis
RAG
mTORC1 Serine PHDGH-amplified
synthesis cancer cell
RHEB Potential for
targeting in
cancer
TSC

Lysosome
AKT
PI3K
Increased activity
and membrane Increased PI3K Macropinocytosis
abundance signalling in cancer

Amino acid RTK


transporter RAS-transformed Extracellular
Growth cancer cell protein
Amino acids factor

Fig. 2 | amino acid sensing and acquisition. Cellular levels of amino acids can be increased by increased expression
and cell membrane retention of amino acid transporters, which happens downstream of receptor tyrosine kinase (RTK)
signalling mediated by growth factors.  In addition, mechanistic target of rapamycin complex 1 (mTORC1) senses growth
factor signalling and the presence of amino acids to promote the utilization of amino acids by activating protein translation.
CASTOR1, Sestrin-2 and SAMTOR are amino acid sensor proteins for arginine, leucine and methionine (through
S-adenosylmethionine (SAM)), respectively. These sensors control the activity of the GATOR2–GATOR1 complexes,
negatively regulating the activity of RAG GTPases and hence inhibiting mTORC1 localization to the lysosome and
activation. The suppressive functions of these sensors are diminished in the presence of the amino acids that they sense,
leading to the activation of mTORC1. When amino acid levels are low , general control nonderepressible 2 (GCN2) is
activated by the increase in uncharged tRNAs and results in the inhibition of global protein translation (not shown).
Paradoxically , GCN2 activation leads to increased expression of activating transcription factor 4 (ATF4). ATF4 mediates an
integrated stress response that includes the upregulation of genes that mediate amino acid uptake, synthesis and utilization
(translation) to eventually support cell adaptation and recovery from stress. Note that ATF4 increases the expression of a
variety of tRNA synthetases that enable the cell to better capture the limited amount of cellular amino acids for protein
translation, which facilitates the production of proteins that are critical in stress adaptation and recovery. Cells can also
acquire amino acids through macropinocytosis of extracellular proteins followed by their lysosomal degradation.
Macropinocytic utilization is inhibited by mTORC1 and is promoted in cancer via oncogenic RAS and the PI3K pathway.
Certain cancers have also been associated with upregulation of phosphoglycerate dehydrogenase (PHGDH), leading to
increased serine synthesis. These cancers depend on PHGDH for growth, and interference with de novo serine biosynthesis
could be explored as a potential strategy in cancer treatment. Cancer-associated adaptations are highlighted in red. ASNS,
asparagine synthetase; PSAT1, phosphoserine aminotransferase 1; RHEB, RAS homologue enriched in brain; TSC, tuberous
sclerosis complex; x(c)(−), cystine–glutamate antiporter.

www.nature.com/nrm
Reviews

protein translation. Paradoxically, during integrated to genetic or pharmacological interference of this path­
stress response, translation initiation can still occur at way54–56. Furthermore, beyond PHGDH-overexpressing
alternative open reading frames, present in certain genes, tumours, targeting serine and glycine in general holds
such as ATF4 (encodes activating transcription factor 4), promise for controlling cancer cell growth, as serine and
leading to the upregulation of their protein products42 glycine metabolism is critically involved in the regu­
(FiG. 2). ATF4 functions as a transcription factor and lation of redox balance — which is often disturbed in
induces the expression of a cohort of genes that together cancer cells — and in support of nucleotide synthesis57
promote cell survival upon metabolic stress. An impor­ (see also below). Restriction of dietary serine and glycine
tant group of genes downstream of ATF4 encodes has been shown to reduce tumour growth in xenograft
proteins that constitute amino acid transporters. For models and in genetically engineered mouse models of
example, expression of both subunits of the x(c)(−) sys­ intestinal cancer and lymphoma58,59.
tem, SLC7A11 (encoding cystine–glutamate antiporter Aspartate is another NEAA that is important for pro­
xCT) and SLC3A2 (encoding common 4F2 heavy chain), liferating cells because it is an essential substrate for
is strongly induced upon ATF4 activation, promoting nucleotide synthesis60 (FiG. 1). However, the extracellu­
cystine uptake at the expense of cellular glutamate (FiG. 2) lar free aspartate level is low, and most cells express the
and maintaining redox homeostasis43–45 (see also below). aspartate transporter SLC1A3 at low levels61. Cellular
In addition, expression of a variety of other amino acid aspartate is mostly derived from the amination of the
transporter genes, including SLC1A5 (encoding ASCT2; TCA cycle intermediate oxaloacetate (FiG. 1; see also
also known as ATB(0)), SLC7A5 (encoding LAT1), Supplementary Box 1), and flux through the TCA cycle
SLC7A1 (encoding CAT1) and SLC6A9 (encoding has been shown to be important in supporting aspartate
GLYT1), is elevated by ATF4, which in turn facilitates synthesis. A major function of the mitochondrial elec­
uptake of amino acids including glutamine, leucine, argi­ tron transport chain (ETC) is to enable access to oxygen
nine, lysine and glycine43,46–48. ATF4 also increases the as the terminal electron acceptor in order to regenerate
expression of almost all cytosolic aminoacyl tRNA syn­ NAD+ and sustain TCA cycle activity. Remarkably, in
thetases, enhancing the capture of the acquired amino cancer cells, reduced cell proliferation caused by ETC
acid for maintaining protein synthesis43,49. In the mean­ inhibition can be rescued with direct aspartate supple­
time, through the induction of Sestrin-2, ATF4 prevents mentation62,63. However, restoration of cell growth by
excessive amino acid consumption by mTORC1 until aspartate upon ETC inhibition does not occur in all
intracellular amino acid levels are restored50 (FiG. 2). cell types. In endothelial cells, ETC inhibition leads to
reduced cell proliferation as well as decreased aspartate
Synthesizing non-essential amino acids. Apart from levels, but aspartate supplementation is not sufficient
direct amino acid uptake, cells are capable of synthe­ to rescue the impaired cell growth caused by ETC defi­
sizing a variety of NEAAs when needed. Many NEAAs ciency64. Moreover, regulatory T cells and haematopoi­
have crucial roles in cell growth beyond serving as etic stem cells are able to proliferate in the absence of
building blocks for protein translation, including acting ETC activity, although the abilities to suppress T cell
as substrates for nucleotide synthesis and supporting function and to initiate erythropoiesis, respectively, are
cellular redox homeostasis (see also below). As a con­ considerably impaired65,66. Nonetheless, these findings
sequence, proliferating cells often demonstrate meta­ highlight that, by supporting aspartate production,
bolic ­adaptations that favour the synthesis of NEAAs. mitochondrial respiration is important for anabolic
However, production of NEAAs generally requires metabolism beyond its conventional ATP-generating
reallocation of nutrients and energy and is therefore role. Because of the dependency of certain cancer cell
­rigorously regulated. types on ETC activity, associated TCA flux and conse­
When cells sense a decrease in amino acid levels quent robust aspartate pro­duction, ETC inhibition and
through GCN2, ATF4 activation can directly lead to the therapeutic aspartate deprivation may be explored as
upregulation of enzymes involved in NEAA synthesis, potential strategies to impede cancer growth67–69.
such as ASNS in asparagine synthesis51 and PHGDH
and PSAT1 in serine biosynthesis52 (FiG. 2). Proliferating Protein scavenging as an amino acid source. As
cancer cells often engage in NEAA production in a described above, cells can specifically adjust the levels
more proactive manner. For example, recurrent genetic of certain amino acids by modulating the expression of
amplifications of the PHGDH gene have been identified selective transporters and/or by regulating de novo
in melanoma and breast tumours, which correlate with biosynthetic pathways. In addition, mammalian cells
increased glycolytic flux into the serine biosynthesis can use a non-selective endocytic pathway, termed
pathway53,54. In addition to serine and glycine produc­ macropinocytosis, to take up nutrients in bulk from
Alternative open reading tion, the transamination reaction in the serine biosynthe­ the extracellular space. Cells initiate macropinocytosis
frames
sis pathway also generates α-ketoglutarate (αKG) from through protrusions of the plasma membrane that fold
Different open reading frames
within a gene, the transcription glutamate, which was shown to be critical for replen­ back to the membrane forming macropinosomes, which
of which can lead to different ishing the TCA cycle, while metabolic intermediates of then are trafficked to fuse with lysosomes for cargo
gene products that may assume the TCA cycle can leave the cycle and act as precursors degradation70,71 (FiG. 2). In principle, cells can recover
different biological roles. for the biosynthesis of macromolecules such as nucleo­ various types of nutrients from macropinocytosis.
Cystine
tides and fatty acids54 (Supplementary Box 1). De novo Macropinocytic scavenging of proteins has been demon­
The oxidized dimer form of the serine synthesis appears essential to cancer cells with strated to be a crucial amino acid source to ­support cell
amino acid cysteine. PHGDH overexpression, as they are exquisitely sensitive proliferation, especially in cancer71,72.

Nature Reviews | Molecular Cell Biology


Reviews

Growth in the AMP-to-ATP ratio and, via its kinase activ­


Glucose factor
ity, restores metabolic homeostasis by suppressing
GLUT1 RTK anabolism and enhancing catabolic processes such as
macro-autophagy (hereafter referred to as autophagy)79.
AKT Protein Beyond its role in promoting lysosomal degradation of
acetylation self-constituents during autophagy, AMPK can also
regulate macropinocytic degradation of extracellular
fluids and necrotic cell debris80. In addition, as a mas­
ter sensor of certain amino acids, mTORC1 acts as a
ACLY ACC negative regulator of macropinocytic trafficking to the
TCA Citrate Acetyl Malonyl-
cycle -CoA CoA lysosome (FiG. 2). Suppression of mTORC1 activity lim­
NADPH its the utilization of extracellular free amino acids but
FASN greatly enhances the catabolism of extracellular pro­
NADP+ teins via macropinocytosis78,81. This paradoxical role of
Mitochondrion
ER stress Palmitate mTORC1 may explain the fact that, while genes encod­
ing RTK, PI3K and PTEN are most frequently altered in
many types of cancer, activating mutations in mTORC1
(O2, Fe2+)
are rare. Constitutive mTORC1 activation would lead
Unsaturated SCD Saturated
fatty acid fatty acid
to increased dependencies on the utilization of extra­
cellular free amino acids and hence would limit the
Fatty acid flexibility of nutrient acquisition strategies in the face
translocase
of ­fluctuations in the extracellular environment during
LPL tumour progression.
Circulating triglycerides
in lipoprotein complexes Elevation of fatty acid synthesis
Fig. 3 | Fatty acid synthesis. Fatty acids can be taken up from the environment (for Fatty acids are the building blocks for all lipids and
example, from the circulation) through the activity of lipoprotein lipase (LPL) and fatty hence are key constituents of all biological membrane
acid translocase.  In addition, fatty acid synthesis can occur de novo. Growth factor structures. As a result, proliferating cells need to acquire
signalling promotes utilization of glucose for fatty acid synthesis via redirecting citrate sufficient fatty acids to support membrane growth
(dashed arrow) away from the tricarboxylic acid (TCA) cycle. AKT activation results in and integrity.
the increase in fatty acid synthesis, partly by promoting the activity of ATP citrate lyase
(ACLY). Acetyl-CoA carboxylase (ACC) and fatty acid synthase (FASN) are both Synthesizing fatty acids. Most normal cells, even when
involved in the early steps of fatty acid synthesis, leading to the formation of palmitate. they are in a proliferative state, take up much of their
These enzymes have been shown to be elevated in cancers and hence are potential
required fatty acids from the circulation via the activity
targets in cancer treatment (highlighted in red boxes). The production of saturated fatty
acids needs to be balanced with that of unsaturated fatty acids to avoid endoplasmic of lipoprotein lipase (LPL) and fatty acid translocases
reticulum (ER) stress, which occurs in response to accumulation of saturated fatty acids in such as CD36 (refs82–84). However, it has been known
cellular (including ER) membranes and consequent disruption of membrane homeostasis. since the 1950s that tumour cells have elevated fatty
This balance is partly achieved through the regulation of the oxygen-dependent and acid synthesis activity, which is further exacerbated as
iron-dependent enzyme, stearoyl-CoA desaturase (SCD). Note that acetyl-CoA acts cancer progresses. De novo fatty acid synthesis makes a
as the substrate for acetylation of histones as well as a variety of non-histone proteins. major contribution to the intracellular fatty acid pool of
This link has been demonstrated to be an important mechanism by which cells tumour cells83–85.
coordinate metabolic status with gene expression and protein activities. GLUT1, glucose Elevated endogenous fatty acid synthesis is sup­
transporter 1; RTK , receptor tyrosine kinase. ported by the increased glycolytic flux observed in most
cancer cells. As glucose carbon enters the TCA cycle,
Macropinocytosis is evolutionarily conserved. Both a considerable amount is diverted from the cycle and
unicellular and multicellular eukaryotes can acquire exported back to the cytosol as citrate (FiG. 3; see also
nutrients through macropinocytosis73. Similar to other Supplementary Box 1). The ATP citrate lyase (ACLY)
nutrient acquisition strategies, mammalian cells regu­ functions to cleave citrate into oxaloacetate while
late macropinocytosis at multiple levels. The initiation releasing acetyl-CoA, which is the primary substrate
of macropinocytosis is characterized by membrane ruf­ for fatty acid chain elongation. Through the activity of
fling and macropinosome formation and is normally acetyl-CoA carboxylase (ACC), acetyl-CoA is converted
stimulated by growth factor signalling and PI3K activa­ into malonyl-CoA and is committed to fatty acid synthe­
tion74,75. Importantly, oncogenic RAS is a major regulator sis. Fatty acid synthase (FASN) carries out the stepwise
of the rate and volume of macropinosome uptake (FiG. 2). condensation of malonyl-CoA, elongating the fatty acid
Macropinosome cargo can sustain the proliferation of chain until palmitate is generated, which serves as the
RAS mutant cancer cells by providing nutrients in poorly precursor for production of various other fatty acid spe­
vascularized tumour regions76–78. cies83 (FiG. 3). Enzymes in the fatty acid synthesis pathway,
As part of the cellular nutrient acquisition net­ FASN in particular, are found to have increased expres­
work, macropinocytosis is coordinated with other sion levels across various cancer types, and this increased
nutrient-sensing and nutrient-responsive pathways expression correlates with worse clinical outcomes86–88.
in a concerted manner. AMP-activated protein kinase In fact, many cancer cells become dependent on the
(AMPK) monitors energy stress by sensing an increase fatty acid synthesis pathway, and targeting key enzymes

www.nature.com/nrm
Reviews

of de novo fatty acid synthesis has emerged as a potential are able to release unsaturated fatty acids into phospho­
therapeutic option in cancer treatment83,89. lipid pools upon hypoxia-induced SCD1 inhibition98.
Upregulated enzymatic activity, together with the Moreover, in certain liver and lung carcinomas, it was
associated cellular metabolic changes, helps to explain suggested that palmitate can be desaturated to produce
how cancer cells achieve increased levels of fatty acid sapienate, thereby acting as an alternative means to gen­
synthesis. It remains controversial what advantages erate unsaturated fatty acids, relieving the dependence
tumour cells obtain by relying more on de novo fatty acid on SCD1 (ref.99).
synthesis, despite their access to environmental lipids.
One possibility is that, by actively elevating fatty acid Production of nucleotides
synthesis and stocking up on carbon and energy, cancer In comparison with other nutrients, nucleotide produc­
cells can better prepare themselves for regional nutrient tion through endogenous mechanisms is of particular
scarcity when the tumour expands or when cancer cells importance to proliferating cells because the amount
experience adverse environments during metastasis and of nucleotides directly taken up from the extracellular
recolonization. For example, metastasis in certain can­ space is negligible. During cell proliferation, the demand
cer types was shown to depend on the ability of tumour for nucleotides increases owing to the need to synthesize
cells to access and metabolize lipids90,91. Moreover, it was ribosomal RNA (rRNA), duplicate the genome (synthe­
found that de novo fatty acid synthesis is essential for size DNA) and maintain the transcriptome (produce
cancer cells to survive antiangiogenic therapy and allows large amounts of mRNA). Nucleotide biosynthesis is
tumour cells to rapidly regrow and metastasize once the also uniquely positioned in the cellular metabolic net­
treatment is withdrawn92. Another potential benefit that work, because pathways that lead to ribose, pyrimidine
cancer cells may gain from fatty acid synthesis is related and purine production require substrate and energy
to the fact that fatty acid synthesis converts a consider­ input from multiple metabolic processes, including the
able amount of the reducing equivalent of NADPH into pentose phosphate pathway, one-carbon-unit cycle, ETC
NADP+, thereby contributing to cellular redox balance and the TCA cycle (FiG. 1; Supplementary Box 1).
by serving as an electron acceptor (see also below). This
was proposed to be particularly important in hypoxic Generating ribose 5-phosphate. Ribose 5-phosphate is
tumour cells where oxygen, a major electron acceptor, a phosphorylated form of the pentose sugar, ribose, on
is limited93. Nonetheless, increased fatty acid synthesis which nitrogenous bases are built to form nucleotides.
may assume different roles under distinct malignant Cellular ribose 5-phosphate is normally derived from the
contexts and during different cancer stages. Further oxidative phase of the pentose phosphate pathway, where
Endoplasmic reticulum (ER) mechanistic delineation of the role of fatty acid synthesis the rate-limiting enzyme glucose-6-phosphate dehydro­
stress in ­supporting cell growth and tumorigenesis is needed. genase (G6PD) diverts glucose away from glycolysis to
Cellular stress in the ER that
often results from the
power ribose 5-phosphate production and NADPH
accumulation of misfolded Balancing saturated and unsaturated fatty acids. In generation (Supplementary Box 1). The phosphoribo­
proteins or a failure to maintain addition to increasing fatty acid synthesis, it is equally syl pyrophosphate synthetase (PRPS) can subsequently
the membrane integrity of the important for proliferating cells to balance the content of convert ribose 5-phosphate into phosphoribosyl pyro­
ER.
saturated and unsaturated fatty acids. Accumulation phosphate (PRPP), which activates ribose 5-phosphate
Lysophospholipids of excess saturated fatty acids is known to trigger and commits it to nucleotide biosynthesis (FiG. 4).
Derivatives of phospholipids endoplasmic reticulum (ER) stress, partly by impairing The production of nucleotides is energy demanding.
in which one of the two acyl ER membrane fluidity94. In mammalian cells, fatty acid For example, during purine synthesis, seven molecules
chains is lost. desaturation is carried out by stearoyl-CoA desaturases of ATP are consumed in order to build a molecule of
Sapienate
(SCDs) that function primarily at the ER, converting inosine monophosphate (IMP) on PRPP, and an addi­
A fatty acid that is usually a satu­rated fatty acids into monounsaturated fatty acids tional molecule of ATP or GTP is required to convert
component of the secretion in an oxygen-dependent and iron-dependent manner IMP into AMP or GMP. As a result, nucleotide synthesis
from sebaceous glands in the (FiG. 3). Given the critical role of SCDs in maintaining must be tightly coordinated with cellular energy status.
skin. Sapienate can be
cellular unsaturated fatty acid levels, development of Indeed, the enzymatic activity of PRPS is diminished as
synthesized from palmitate
by desaturation. inhibitors to SCD1, the major form of SCDs in most ADP and GDP levels increase, which reflects the accu­
human cells, has been an active area of research in treat­ mulation of nucleotide products, as well as indicating a
Pentose phosphate pathway ing cancer as well as metabolic syndromes associated drop in cellular energy level100. Moreover, it was found
A metabolic pathway parallel with increased lipid accumulation95. that AMPK activation upon energy stress can directly
to glycolysis that involves the
generation of various pentoses
Because of the dependence on oxygen for desatura­ phosphorylate PRPS and convert the enzyme from its
including ribose 5-phosphate tion reactions, the activity of SCD1 is strongly reduced active hexamer form into the inactive monomer form101
that functions as a precursor under hypoxic conditions94,96. Because the progression (FiG. 4). These mechanisms reduce nucleotide synthesis
for nucleotide synthesis. of tumours often involves periods of oxygen limita­ when the cellular energy status is not optimal.
tion, cancer cells need to employ alternative means to
One-carbon-unit cycle
A group of biochemical acquire unsaturated fatty acids and therefore become less Building pyrimidine nucleotides. Pyrimidine
reactions that involve the dependent on SCD1. For example, it was shown that cells nucleotide synthesis is initiated in the cytosol by
transfer of the one-carbon transformed by oncogenic RAS have enhanced ability carbamoyl-phosphate synthetase-II (CPS-II), which
groups between various to scavenge extracellular lysophospholipids to supple­ utilizes ATP, HCO3− and the amide group of glutamine
molecules such as
tetrahydrofolate,
ment cellular unsaturated fatty acids under hypoxia or to form carbamoyl phosphate. Through the activity
S-adenosylmethionine and SCD1 inhibition97. In the context of clear-cell renal cell of aspartate transcarbamoylase (ATCase), carbamoyl
vitamin B12. carcinoma, triglycerides stored in cellular lipid droplets phosphate is then condensed with aspartate to produce

Nature Reviews | Molecular Cell Biology


Reviews

Pyrimidine biosynthesis

HCO3– Aspartate Glutamine


Glutamine Dihydroorotate Orotate Uracil Cytosine
CAD O NH2
C
5,10-CH2-THF
DHODH C
HN C N C
Growth factor mTORC1 e– H2O
signalling QH2 C C
Thymine C C
O N O N
O2 H O H
Coupling to amino Coupling to C CH3
acid homeostasis ETC and energy ATP HN C
metabolism Mitochondrion
C C
O N
Purine biosynthesis H
Coupling NH2
to energy mTORC1
C N
Glucose homeostasis N C
C
C C
N N
AMPK
Folate Adenine O
Ribose-5-P Aspartate C N
HN C
PRPS C
N10-formyl-THF HCO3– N10-formyl-THF Glutamine C C
H2N N N
PRPP RPA IMP Guanine
IMPDH
Glutamine Glycine Glutamine Aspartate

Fig. 4 | Synthesis of pyrimidine and purine nucleotides. Nucleotide synthesis is key for cell proliferation as it is required
for DNA replication, gene transcription and ribosome biogenesis, and the uptake of nucleotides from extracellular sources
is negligible.  Nucleotide biosynthesis interrogates multiple metabolic pathways and requires the utilization of various
substrates as the carbon and nitrogen sources, including amino acids and metabolites of the folate cycle (the colours of
carbon and nitrogen in the chemical structure of pyrimidines and purines correspond to the contributing source).
Pyrimidine synthesis intersects with amino acid metabolism via positive regulation of carbamoyl-phosphate synthetase II,
aspartate transcarbamoylase and dihydroorotase (CAD) downstream of mTROC1 as well as with ATP production and
energy metabolism (electron transport chain (ETC) via the activity of dihydroorotate dehydrogenase (DHODH), highlighted
in red, which has been implicated as a potential target for cancer treatment). Purine synthesis is indirectly regulated by
mechanistic target of rapamycin complex 1 (mTORC1) via its stimulation of the mitochondrial folate cycle. In addition,
the key energy sensor AMP-activated protein kinase (AMPK) negatively regulates purine biosynthesis by inhibiting the
activity of phosphoribosyl pyrophosphate synthetase (PRPS). This enables fine-tuning of nucleotide synthesis in accordance
with cellular energy metabolism. IMPDH, inosine monophosphate dehydrogenase; PRPP, phosphoribosyl pyrophosphate;
QH2, dihydroxyquinone; Ribose-5-P, ribose 5-phosphate; RPA , phosphoribosyl amine; THF, tetrahydrofolate.

carbamoyl aspartate. Dihydroorotase subsequently circu­ DHODH instead transfers electrons to ubiquinone, which
larizes carbamoyl aspartate to form the ring-structured then passes the electrons on to respiratory complex III
dihydroorotate. In human cells, CPS-II, ATCase and and ultimately to oxygen106. As a result, the production
dihydroorotase activities are all contained in the tri­ of orotate in pyrimidine synthesis is coupled to the
functional protein CAD (FiG. 4). Consistent with the ETC and energy generation, which exerts an additional
hypo­thesis that normal cell growth is dependent upon control over nucleotide synthesis and ensures that it is
growth factor stimulation, CAD is known to be regulated coupled to cellular energy status. The unique metabolic
by growth factor signalling, partly through the action of positioning of this reaction also makes DHODH a vulner­
MAPK102,103. Furthermore, downstream of mTORC1, able node within pathways of growth regulation that
S6K1 was found to directly phosphorylate CAD and to has the potential to be a cancer treatment target. In line
induce its activity to provide pyrimidine nucleotides for with this, it was found that combinatorial inhibition of
ribosomal biogenesis, hence supporting protein transla­ oncogenic BRAF-V600E (BRAF proto-oncogene, V600E
tion104,105. By positioning CAD under the regulation of mutation) and DHODH can lead to a reduction in mela­
mTORC1, the initiation of pyrimidine synthesis is only noma tumour growth107. Similarly, high-throughput
favoured in the presence of sufficient amino acid sub­ screening identified DHODH as a drug target, which
strates, as monitored by mTORC1 activity (FiG. 4). As a when inhibited induces differentiation of a wide range
result, nucleotide production is coordinated with amino of acute myeloid leukaemia cells108.
acid availability, which together support cell proliferation. Subsequently, to complete pyrimidine synthesis,
Ubiquinone A unique feature of the next step in pyrimidine syn­ orotate is combined with PRPP to form uridine mono­
A coenzyme that functions as thesis is that the enzyme dihydroorotate dehydrogenase phosphate (UMP) through the activity of uridine
an electron carrier in various (DHODH) is localized on the outer surface of the inner monophosphate synthetase (UMPS). All other pyrimi­
biological processes, including
the electron transport chain as
mitochondrial membrane, oxidizing dihydroorotate to dine nucleotide species are derived from UMP. Notably,
part of aerobic cellular produce orotate (FiG. 4). Unlike many other dehydro­ in order to produce cytidine triphosphate (CTP), an
respiration. genases that require NAD+ or NADP+ as electron acceptor, additional molecule of glutamine is required to provide

www.nature.com/nrm
Reviews

Folate cycle
the amide group (FiG. 4). Moreover, for DNA synthe­ and aspartate nitrogen join to form phosphoribosyl­
A group of biochemical sis, deoxy UMP (dUMP) is converted into deoxy­ aminoimidazole succinocarboxamide, which is further
reactions occurring in both the thymidine monophosphate (dTMP) by thymidylate cleaved by adenylosuccinate lyase to release amino­
cytosolic and mitochondrial synthase (encoded by TYMS). This reaction requires the imidazole carboxamide ribonucleotide (AICAR) and the
compartments that involve the
folate cycle intermediate 5,10-methylene-tetrahydro­ TCA cycle intermediate fumarate. Finally, the common
metabolism of folate and its
congeners. The folate cycle folate (5,10-methylene-THF) as the source of the purine precursor IMP is generated by AICAR formyl­
can provide critical metabolic methyl group (FiG. 4) and is a target for the antimetabolite transferase, IMP cyclohydrolase, with the addition of a
intermediates for the 5-fluorouracil in cancer treatment109. second one-carbon unit from N10-formyl-THF.
biosynthesis of macromolecules
The other species of purine nucleotides are derived
such as nucleotides.
Assembling purine nucleotides. Synthesis of purine from IMP. To synthesize AMP, the adenylosuccinate syn­
Antimetabolite nucleotides also utilizes various carbon and nitrogen thase takes an additional molecule of aspartate, and the
A chemical used in cancer sources and integrates a number of metabolic path­ resulting adenylosuccinate is cleaved to release AMP and
treatment that often interferes ways (FiG. 4). The entire purine synthesis pathway takes fumarate. Production of GMP involves first the oxida­
with cellular nucleotide
synthesis and DNA replication.
place in the cytosol. PRPP-amidotransferase (PPAT) tion of IMP by IMP dehydrogenase (IMPDH) to form
first displaces the pyrophosphate group in PRPP by the xanthosine monophosphate (XMP) and subsequently
5-Fluorouracil amide group from glutamine to produce phosphoribo­ the amination of XMP to GMP using the amide group
A medication used in cancer syl amine. Phosphoribosyl amine is then converted into from glutamine.
treatment primarily by
the ring-structured phosphoribosylaminoimidazole, The production of purine nucleotides requires
targeting the thymidylate
synthase to block thymidine
utilizing carbon and nitrogen from glycine, glutamine the direct contribution of carbon and nitrogen from
synthesis. and N10-formyl-THF. Subsequently, the HCO3− carbon glycine and N10-formyl-THF (FiG. 4). As a result, the
folate cycle and the metabolism of serine and glycine
are intimately connected to the regulation of purine
Growth factor synthesis. In human cells, folate metabolism occurs in
Low O2 RTK both the cytosolic and mitochondrial compartments.
Angiogenesis
When studied in cell culture, the mitochondrial serine
HIF1α
hydroxymethyltransferase 2 (SHMT2) favours the gen­
Lysophospholipids
Macropinocytosis
GLUT1 eration of glycine from serine, while transferring the
Glucose β-carbon of serine to tetrahydrofolate (THF) to produce
VEGF
Extracellular 5,10-methylene-THF. Methylene-THF dehydrogenase 2
protein (MTHFD2) subsequently catalyses the production of
Fatty acids Lactate MCT
Amino acids
N10-formyl-THF, which is mostly exported to the cyto­
Biosynthesis sol in the form of formate. The cytosolic arm of folate
and energy metabolism then regenerates N10-formyl-THF from
metabolism
formate to support purine biosynthesis57. Similar to
Regulatory ECM utilization
M2 pyrimidine synthesis, purine production is also highly
T cell macrophage coordinated with the cellular nutrient status and growth
(integrin-mediated Amino
uptake) ECM signalling pathways. For example, mTORC1 activity
acids
was demonstrated to promote purine synthesis, partly
through the regulation of enzymes involved in serine
Autophagy biosynthesis and folate metabolism110.
TGFβ Fibroblast TGFβ Taken together, these findings show that, in order to
build a full battery of pyrimidine and purine nucleotides,
Fig. 5 | Metabolic interactions with the extracellular environment. Extensive metabolic cells need a continuous supply of carbon and nitrogen
interactions exist between proliferating cells and the microenvironment. In the case of derived from a variety of amino acids and metabolic
cancer cells and the tumour microenvironment, cancer cells receive metabolic signalling intermediates. The nucleotide synthesis pathways are
cues such as those from growth factors, which can be provided by infiltrating immune intimately coupled with multiple metabolic processes.
cells. These cues rewire their metabolic programmes to support biosynthesis and growth
Because of these extensive metabolic interactions,
(FiGs 1–4). Hypoxia (low O2 levels), which is a hallmark of solid tumours, further promotes
metabolic rewiring by activating hypoxia-inducible factor 1α (HIF1α), which, among other nucleo­tide synthesis is under rigorous cellular control.
functions, promotes aerobic glycolysis and lactate production. Lactate secreted by cancer Many of the antimetabolites currently used in cancer
cells can have effects on the surrounding environment. For example, cancer-cell-derived therapy exploit the process of nucleotide biosynthesis as
lactate has been shown to induce secretion of vascular endothelial growth factor (VEGF), a way to selectively impair proliferating cells.
thereby promoting angiogenesis and increasing the potential of cancer dissemination.
Cancer cells also acquire nutrients directly from the environment, including glucose, Interactions with microenvironment
amino acids and macromolecules, such as components of the extracellular matrix (ECM) Beyond the cellular metabolic network, extensive meta­
via integrin-receptor-mediated endocytosis as well as other extracellular proteins bolic interactions also occur between a proliferating cell
and lysophospholipids via macropinocytosis. Specialized fibroblasts, known as and the extracellular environment in which it resides.
cancer-associated fibroblasts (CAFs), have a particularly important role in cancer. CAFs
Multiple components are present in this microenviron­
have the ability to support cancer cell growth by mechanisms involving ECM deposition
and autophagy. Autophagy in CAFs and other stromal cells present in the cancer ment, including the vasculature, extracellular matrix
environment can result in the release of nutrients, such as amino acids, which can be taken (ECM) and various other cell types such as fibroblasts
up and utilized by cancer cells (for example, as building blocks for protein synthesis or for and immune cells (FiG. 5). The interactions between
bioenergetics). GLUT1, glucose transporter 1; MCT, monocarboxylate transporter ; RTK , proliferating cells and their surroundings are bidirec­
receptor tyrosine kinase; TGFβ, transforming growth factor-β. tional and multifaceted. Here, we consider how the

Nature Reviews | Molecular Cell Biology


Reviews

extracellular environment influences the metabolism of The resultant increase in lactate production, in turn, was
proliferating cells. shown to enhance VEGF secretion by immune cells and
to stimulate angiogenesis to supply nutrients and oxygen
Receiving metabolic stimulation from the niche. for the progressive development of tumours as they grow
Wound healing is a paradigm process in which cell in size122. Overall, cancer cells harness properties of the
proliferation is enhanced in a physiological context. wound-healing process during prolonged interaction
A healing wound represents a typical milieu in which with the TME to adjust their metabolic programmes in
crosstalk occurs between the parenchymal cells that support of proliferation.
must expand in number to repair damaged tissue and
the associated stromal and immune cells that provide Acquiring nutrients from opportunistic sources. To
environmental support. Wound healing is a highly coor­ support the high rates of proliferation, transformed
dinated process that involves complex metabolic inter­ cancer cells often deplete their microenvironment of
actions between the cell types involved. The process is a variety of nutrients, such as NEAAs, that are critical
initiated by inflammation at the injury site, which is in for anabolic growth123,124. As a result, cancer cells often
part achieved when innate immune cells are recruited need to turn to opportunistic modes of nutrient acqui­
by the damage-associated molecular patterns — including sition from the TME to maintain tumour growth and to
uric acid, nucleotides and hyaluronan fragments — that adapt to the changes in their environment. As discussed
are released as a consequence of tissue damage111–113. previously, RAS-transformed cancer cells are able to
These molecules act as signals to elicit inflammatory scavenge extracellular protein and lysophospholipids
responses preceding tissue repair114. During epithelial to use as a source of amino acids and fatty acids77,97. In
regeneration, the enrichment of growth factors released addition to utilizing extracellular fluids, cancer cells are
by immune cells and fibroblasts into the wound bed able to catabolize ECM components deposited by fibro­
serves as a signal to allow nutrient uptake and prolif­ blasts, such as laminin and collagen, which are present
eration by the remaining cells to expand and establish in abundance in the TME, to retrieve amino acids under
re-epithelialization. Meanwhile, immune-cell-derived nutrient starvation125,126 (FiG. 5).
transforming growth factor-β (TGFβ) induces cell In contrast to proliferating cells, most stromal cells
fate change in local fibroblasts, which in turn produce in the TME turn to autophagic degradation of intra­
and deposit ECM proteins to support tissue remodel­ cellular constituents to survive nutrient limitation.
ling114,115. Tissue damage is also often associated with Counterintuitively, autophagic cells release some of
disruptions of blood supply and oxygen delivery. The their recycled metabolic intermediates into the TME.
local hypoxic environment stimulates the production of This release of nutrients can in turn support the growth
VEGF by macrophages, which promotes endothelial cell of neighbouring cancer cells127 (FiG. 5). For example,
proliferation for neovascularization114,116 (FiG. 5). in the context of pancreatic ductal adenocarcinoma,
The development of a tumour resembles the process stroma-associated pancreatic stellate cells release ala­
of wound healing in many aspects. In fact, it was pro­ nine as a major product of autophagy, and this released
posed that tumours can be studied as ‘wounds that do alanine can fuel the TCA cycle in pancreatic ductal
not heal’116. From the metabolic perspective, interactions ­adenocarcinoma cells128.
between cancer cells and the tumour microenvironment Beyond taking up catabolic products from stromal
(TME) not only co-opt features of wound healing but cells, cancer cells may adapt to actively utilize a variety
also manifest novel characteristics, which likely result of surrounding nutrients, particularly during metasta­
from the heterogeneous nature of oncogenic transfor­ sis, as they invade new territories that are metabolically
mation and the stromal environment of the organ in challenging. For example, omentum invasion is a major
Damage-associated which a tumour arises. For example, the presence of form of ovarian cancer metastasis. It was suggested that
molecular patterns hyaluronan in the TME was shown to serve as a meta­ upregulation of fatty acid-binding protein 4 (FABP4) in
A group of biomolecules bolic instruction that promotes glycolysis in cancer ovarian cancer cells is one of the determinants of their
that are enriched upon
cells by dampening thioredoxin -interacting protein homing at the omentum as it enables cancer cells to
non-infectious inflammatory
responses such as during the (TXNIP) levels, which promotes GLUT1 internalization, acquire lipids directly from omental adipocytes — in
process of wound healing. thereby enriching GLUT1 at the plasma membrane117. this case, omental adipocytes serve as an important
Cancer-associated fibroblasts (CAFs) have long been source of energy and fatty acids to support growth of
Thioredoxin recognized to have a deterministic role during tumour metastases129.
A class of small proteins
encoded by the TXN and TXN2
progression118,119. CAFs undergo substantial metabolic
genes that mainly function changes in response to growth factors and the hypoxic Adaptations to metabolic stress
as cellular antioxidants. environment and support the anabolic growth of cancer Cell growth and proliferation expose cells to a new set
cells115. One recent example indicates that, upon TGFβ of stresses associated with meeting the supply demands
Cancer-associated
signalling, the p38 MAPK pathway is activated in CAFs, of anabolic growth while retaining sufficient bioener­
fibroblasts
(CAFs). Cells derived from which promotes cytokine production and release from getics and redox regulation to maintain viability. In addi­
normal fibroblasts within the CAFs to stimulate the use of glycogen stored in adja­ tion, despite extensive metabolic rewiring, proliferating
tumour microenvironment that cent cancer cells as a source of glucose120. In addition, cells are constantly encountering metabolic stresses. This
promote cancer development. the hypoxic environment in the tumour niche signals is particularly true for cancer cells, as their proliferation
Omentum
to cancer cells to reinforce aerobic glycolysis through exceeds the ability of the existing vasculature to supply
Layers of peritoneum that stabilization of hypoxia-inducible factor 1α (HIF1α) sufficient oxygen and nutrients to support both growth
surround abdominal organs. and induction of its downstream gene targets121 (FiG. 5). and survival.

www.nature.com/nrm
Reviews

Surviving nutrient limitations. When nutrient supply accepted by oxygen via ETC in the mitochondria to
is compromised, mammalian cells are able to engage a drive ATP production. Whether glycolytic products
number of mechanisms in order to sustain bioenergetics are catabolized in the mitochondrial TCA cycle or
and support viability. Autophagy is a critical catabolic diverted into anabolic synthesis, the cells are left with
process whereby cells salvage intracellular constituents NADH that must be reconverted into NAD+ to main­
under conditions of nutrient scarcity. Nutrient limita­ tain redox homeostasis. Just as in resting cells, in cancer
tions often result in a reduction in mTORC1 activity and cells with a high glycolytic rate as a result of PI3K muta­
its dissociation from the unc-51-like autophagy activat­ tion, the malate–aspartate shuttle is essential to promote
ing kinase (ULK) complex. As a consequence, the ULK the translocation of cytosolic electrons into the mito­
complex becomes activated and initiates the formation chondria for consumption in oxidative phosphoryla­
of the double-membrane autophagosomes. Through the tion140,141. The malate–aspartate shuttle, therefore, serves
coordination of a series of ATG proteins, the autophago­ as a mechanism to maintain redox homeostasis in the
some membranes encapsulate cargo and deliver the sub­ cytosolic compartment to enable NAD+ regeneration
strates to the lysosome for degradation130,131. Autophagy and its continued supply to support constant glucose
can also be a selective process that involves a variety of utilization (FiG. 6).
cargo receptors that specifically target distinct substrates Not all nutrient usage in proliferating cells is for ana­
in a signal-dependent manner. Selective autophagic bolic synthesis. For example, many cancer cells were
substrates include proteins, lipids and ribosomes130–132. shown to have an increased demand for serine57. Apart
As a result, autophagy pathways repurpose the limited from the anabolic role of serine in protein translation and
intracellular resources and enable cells to survive nutri­ nucleotide synthesis, it was shown that the catabolism
ent and energy crisis. In addition to the recycling of of serine in mitochondria is critical to support NADPH
nutrients, autophagy is also an important mechanism production and redox balance142. In addition, besides its
that mediates the degradation of selected (for example, role as a TCA cycle anaplerotic substrate, glutamine is
damaged or superfluous) organelles to maintain cellular important in maintaining the cellular glutathione pool
integrity and functionality130,131. for redox regulation (see next paragraph), by providing
Autophagy is typically associated with the inhibition glutamate and sustaining the uptake of extracellular
of cell growth. Notably, however, the role of autophagy ­cystine143,144 (FiG. 6).
in cancer progression is context-dependent. On the one As a result of their high glycolytic rate and the pro­
hand, it was suggested in mouse models that autophagy gressive depletion of oxygen in the local environment
deficiency results in increased susceptibility to cancer as the cell mass accumulates, proliferating cells face the
initiation. It is worth noting that a defect in autophagy problem of an elevated NADH-to-NAD+ ratio, which
can lead to genomic instability as well as the activation results in redox imbalance and consequently an increase
of oncogenic signalling cascades, and hence, auto­ in the generation of reactive oxygen species (ROS).
phagy may limit tumorigenesis through mechanisms The mitochondrial ETC is one of the main sources of
that are not directly linked to metab­olism133. On the cellular ROS (FiG. 6). When electrons are being passed
other hand, the nutrient-scavenging function of auto­ to oxygen, a fraction of them escape from the ETC and
phagy has been increasingly associated with its role in directly react with oxygen to produce superoxide, which
sustaining cancer cell growth and tumour progression. is the precursor of many other ROS such as hydrogen
For example, in a mouse model of RAS-driven NSCLC, peroxide and hydroxyl radicals 145. Another source
autophagy was shown to have a critical role in main­ of ROS are the NADPH oxidases (NOXs), which are
taining the cellular nucleotide pool and energy balance membrane-bound enzymes that catalyse the production
to support cancer cell survival134,135. Pancreatic cancer of a superoxide free radical by transferring one electron
cells were found to have elevated basal autophagy acti­ to oxygen from NADPH (FiG. 6). In order to prevent ROS
vity when compared with normal pancreatic cells, and accumulation, which can lead to irreversible damage to
autophagy was demonstrated to be a required compo­ cellular components, cells possess multiple antioxidant
nent for pancreatic tumour growth136–138. In addition, mechanisms such as the thioredoxin system and the
Malate–aspartate shuttle as discussed above, cancer cell-extrinsic autophagy glutathione system. Furthermore, when the ROS level
A biochemical system that pathways — occurring in the host liver or in stromal rises, the master regulator of antioxidant response,
translocates electrons from the cells in the tumour microenvironment — can also nuclear factor erythroid 2 like 2 (NRF2), is stabilized
cytosol into the mitochondria affect the nutrient status and hence the survival and as its negative regulator Kelch-like ECH-associated
for oxidative phosphorylation;
the process involves the
growth of cancer cells128,139. Understanding the role protein 1 (KEAP1) is oxidized and loses its ability to
exchange of malate and of autophagy in supporting tumour cell growth and sequester NRF2 in the cytosol for proteasomal degra­
aspartate between the two proliferation has the potential to reveal metabolic dation. Nuclear NRF2 acts as a transcription factor that
compartments. vulnerabilities and provide new therapeutic options in activates a collection of genes involved in the synthesis
cancer treatments. and utilization of cellular antioxidant systems to clear
Glutathione
A tripeptide of glutamate, excessive cellular ROS.
cysteine and glycine that, Maintaining redox homeostasis. Maintaining redox Interestingly, a modest increase in ROS level can
together with glutathione- homeostasis is essential to all metabolic states. One stimulate cell growth, partly because hydrogen perox­
utilizing enzymes, functions important indicator of cellular redox balance is the ide can oxidize cysteine residues of a number of protein
to maintain cellular redox
homeostasis and to detoxify
maintenance of the NADH-to-NAD+ ratio. In mammal­ phosphatases including PTEN and MAPK phosphatases,
electrophilic compounds such ian cells, electrons can be captured from fuels, such as and inhibit their activities 146. A physiological level
as reactive oxygen species. glucose, converting NAD+ into NADH, and eventually of ROS is essential for many biological processes.

Nature Reviews | Molecular Cell Biology


Reviews

Lactate
MCT
Glucose
Pentose phosphate
NAD+ pathway
High glycolytic NADP+ NADPH
flux
NAD+ NADH NOXs
NADH
Lactate Pyruvate NADP+ NADPH
Maintenance of ROS
redox homeostasis NADH NAD
+ (oxidative stress)
Folate
Aspartate Malate pathway
Glutathione
Glutathione
Malate–aspartate Q synthesis
shuttle NADP +
NADPH I II III C
IV
N10-formyl-THF
Aspartate Malate NADH NAD+ V
TCA Serine
NADH NAD+ cycle

Cystine
Mitochondrion
Glycine x(c)(–)
Glutamine Glutamate

Fig. 6 | Maintaining redox homeostasis in proliferating cells. Owing to a high glycolytic rate and associated biosynthetic
reactions, proliferating cells face the problem of an elevated NADH-to-NAD+ ratio, and thus, maintaining redox homeostasis
is critical during cell proliferation.  Mechanisms to maintain the NADH-to-NAD+ ratio include the malate–aspartate shuttle,
which regenerates cytosolic NAD+ to sustain constant flow through biosynthetic pathways. In addition, an increase in lactate
production supports regeneration of NAD+. At the same time, proliferating cells face the problem of increased generation
of reactive oxygen species (ROS), primarily derived from the electron transport chain in mitochondria and NADPH oxidases
(NOXs). Excessive ROS cause oxidative stress, which curbs cell proliferation and viability. Thus, proliferating cells require
efficient antioxidant systems to counteract the destructive activity of ROS. The two key cellular antioxidant defence
systems include the thioredoxin system (not shown) and the glutathione system. Glutathione is synthesized from glutamate,
cysteine and glycine. Glutamine importantly regulates this process by supplying glutamate and promoting cystine uptake.
Additionally , NADPH is required for efficient ROS detoxification as it is involved in generating reduced glutathione — which
is active against ROS — from its oxidized form (not shown). NADPH is supplied by the conversion of glycolysis intermediates
in the pentose phosphate pathway as well as by mitochondrial serine metabolism in the folate pathway. Antioxidant defence
has been shown to be important for cancer cell survival and tumour growth. For example, cancer cells were demonstrated to
rely on glutathione synthesis and to upregulate mitochondrial folate pathway (highlighted in red). MCT, monocarboxylate
transporter ; THF, tetrahydrofolate; x(c)(−), cystine–glutamate antiporter.

For example, ROS generation by ETC complex III is been shown to undergo reversible metabolic reprogram­
indispensable during the differentiation of mesenchy­ ming to enhance antioxidant capability by modulating
mal stem cells into adipocytes147. ROS that originate the mitochondrial folate pathway to overcome this
from mitochondrial metabolism are also required to barrier152. Targeting the antioxidant system has become
promote cell proliferation and mediate tumorigenesis an area of intense research in cancer biology.
in KRAS-driven cancer cells148. However, excessive ROS
generation can exceed physiological cellular antioxidant Conclusions and perspectives
capacity. Proliferating cells frequently upregulate their Studies over the past decades have greatly advanced our
antioxidant defence mechanisms. In cancer cells, this understanding of cellular metabolism. Metabolic regu­
is most clearly demonstrated by the frequent genomic lation is critically involved in various cellular processes,
alterations in the KEAP1–NRF2 axis that lead to including cell growth and proliferation. Through inves­
increased antioxidant defence and are associated with a tigations of physiological as well as pathological cell pro­
poor cancer prognosis18. Increased antioxidant capacity liferation, the knowledge of how cells acquire and utilize
resulting from NRF2 activation, as well as other mecha­ various types of nutrients for anabolic growth has been
nisms, was found to promote tumorigenesis in various greatly expanded. With this growing knowledge of ana­
cancer settings149–151. Furthermore, oxidative stress has bolic meta­bolism, we have also seen the development
been demonstrated to be a major inhibitory factor for of metabolic interventions in disease treatment. In can­
cancer metastasis. Successful metastatic cancer cells have cer clinics, the metabolic characteristics of cancer cells

www.nature.com/nrm
Reviews

are now used to both diagnose and stage many com­ are components of chemotherapies that are applied
mon tumours, as exemplified through the widespread to the treatment of various types of malignancy154.
use of FDG–PET scanning in cancer evaluation. The Metabolic interventions have had a long and successful
use of antimetabolites as well as the development of history in cancer treatment, and these approaches are
small-molecule inhibitors targeting metabolic enzymes only likely to expand in the coming years.
has led to improved treatment outcomes153. For example,
the nucleoside analogues gemcitabine and cytarabine Published online xx xx xxxx

1. Galdieri, L., Mehrotra, S., Yu, S. & Vancura, A. 23. Lawrence, M. S. et al. Discovery and saturation by regulating ATF4 and xCT expression. J. Biol. Chem.
Transcriptional regulation in yeast during diauxic shift analysis of cancer genes across 21 tumour types. 284, 1106–1115 (2009).
and stationary phase. OMICS 14, 629–638 (2010). Nature 505, 495–501 (2014). 46. Lopez, A. B. et al. A feedback transcriptional
2. Marijuan, P. C., Navarro, J. & del Moral, R. On 24. Pavlova, N. N. & Thompson, C. B. The emerging mechanism controls the level of the arginine/lysine
prokaryotic intelligence: strategies for sensing the hallmarks of cancer metabolism. Cell Metab. 23, transporter cat-1 during amino acid starvation.
environment. Biosystems 99, 94–103 (2010). 27–47 (2016). Biochem. J. 402, 163–173 (2007).
3. Schaller, G. E., Shiu, S. H. & Armitage, J. P. 25. Almuhaideb, A., Papathanasiou, N. & Bomanji, J. 47. Nicklin, P. et al. Bidirectional transport of amino acids
Two-component systems and their co-option for 18F-FDG PET/CT imaging in oncology. Ann. Saudi regulates mTOR and autophagy. Cell 136, 521–534
eukaryotic signal transduction. Curr. Biol. 21, Med. 31, 3–13 (2011). (2009).
R320–R330 (2011). 26. Vander Heiden, M. G., Cantley, L. C. & Thompson, C. B. 48. Zhang, N. et al. Increased amino acid uptake supports
4. DeBerardinis, R. J. et al. Beyond aerobic glycolysis: Understanding the Warburg effect: the metabolic autophagy-deficient cell survival upon glutamine
transformed cells can engage in glutamine metabolism requirements of cell proliferation. Science 324, deprivation. Cell Rep. 23, 3006–3020 (2018).
that exceeds the requirement for protein and 1029–1033 (2009). 49. Shan, J. et al. The C/ebp-Atf response element (CARE)
nucleotide synthesis. Proc. Natl Acad. Sci. USA 104, 27. Faubert, B. et al. Lactate metabolism in human lung location reveals two distinct Atf4-dependent,
19345–19350 (2007). tumors. Cell 171, 358–371 (2017). elongation-mediated mechanisms for transcriptional
5. Rheinwald, J. G. & Green, H. Epidermal growth factor 28. Hui, S. et al. Glucose feeds the TCA cycle via induction of aminoacyl-tRNA synthetase genes in
and the multiplication of cultured human epidermal circulating lactate. Nature 551, 115–118 (2017). response to amino acid limitation. Nucleic Acids Res.
keratinocytes. Nature 265, 421–424 (1977). Faubert et al. (2017) and Hui et al. (2017) provide 44, 9719–9732 (2016).
6. Martin, P. Wound healing—aiming for perfect skin the first evidence that lactate in the circulation can 50. Ye, J. et al. GCN2 sustains mTORC1 suppression
regeneration. Science 276, 75–81 (1997). act as the major carbon source to fuel the TCA upon amino acid deprivation by inducing Sestrin2.
7. Nissen, N. N. et al. Vascular endothelial growth factor cycle in vivo. Genes Dev. 29, 2331–2336 (2015).
mediates angiogenic activity during the proliferative 29. Palm, W. & Thompson, C. B. Nutrient acquisition 51. Siu, F., Bain, P. J., LeBlanc-Chaffin, R., Chen, H. &
phase of wound healing. Am. J. Pathol. 152, strategies of mammalian cells. Nature 546, 234–242 Kilberg, M. S. ATF4 is a mediator of the nutrient-
1445–1452 (1998). (2017). sensing response pathway that activates the human
8. Saltiel, A. R. & Kahn, C. R. Insulin signalling and the 30. Saxton, R. A. & Sabatini, D. M. mTOR signaling in asparagine synthetase gene. J. Biol. Chem. 277,
regulation of glucose and lipid metabolism. Nature growth, metabolism, and disease. Cell 168, 960–976 24120–24127 (2002).
414, 799–806 (2001). (2017). 52. DeNicola, G. M. et al. NRF2 regulates serine
9. Frauwirth, K. A. et al. The CD28 signaling pathway 31. Inoki, K., Li, Y., Zhu, T., Wu, J. & Guan, K. L. TSC2 is biosynthesis in non-small cell lung cancer. Nat. Genet.
regulates glucose metabolism. Immunity 16, phosphorylated and inhibited by Akt and suppresses 47, 1475–1481 (2015).
769–777 (2002). mTOR signalling. Nat. Cell Biol. 4, 648–657 (2002). 53. Locasale, J. W. et al. Phosphoglycerate dehydrogenase
10. Esensten, J. H., Helou, Y. A., Chopra, G., Weiss, A. & 32. Manning, B. D., Tee, A. R., Logsdon, M. N., Blenis, J. diverts glycolytic flux and contributes to oncogenesis.
Bluestone, J. A. CD28 costimulation: from mechanism & Cantley, L. C. Identification of the tuberous sclerosis Nat. Genet. 43, 869–874 (2011).
to therapy. Immunity 44, 973–988 (2016). complex-2 tumor suppressor gene product tuberin as 54. Possemato, R. et al. Functional genomics reveal that
11. Barthel, A. et al. Regulation of GLUT1 gene a target of the phosphoinositide 3-kinase/akt pathway. the serine synthesis pathway is essential in breast
transcription by the serine/threonine kinase Akt1. Mol. Cell 10, 151–162 (2002). cancer. Nature 476, 346–350 (2011).
J. Biol. Chem. 274, 20281–20286 (1999). 33. Menon, S. et al. Spatial control of the TSC complex Locasale et al. (2011) and Possemato et al. (2011)
12. Rathmell, J. C. et al. Akt-directed glucose metabolism integrates insulin and nutrient regulation of mTORC1 provide early evidence that the serine synthesis
can prevent Bax conformation change and promote at the lysosome. Cell 156, 771–785 (2014). pathway is upregulated in cancer and that it is
growth factor-independent survival. Mol. Cell. Biol. 34. Long, X., Lin, Y., Ortiz-Vega, S., Yonezawa, K. & critical in mediating tumorigenesis.
23, 7315–7328 (2003). Avruch, J. Rheb binds and regulates the mTOR kinase. 55. Pacold, M. E. et al. A PHGDH inhibitor reveals
This work demonstrates that growth factor Curr. Biol. 15, 702–713 (2005). coordination of serine synthesis and one-carbon unit
signalling is necessary for mammalian cells to 35. Edinger, A. L. & Thompson, C. B. Akt maintains cell size fate. Nat. Chem. Biol. 12, 452–458 (2016).
maintain glucose uptake in support of and survival by increasing mTOR-dependent nutrient 56. Mullarky, E. et al. Identification of a small molecule
bioenergetics and cell survival. uptake. Mol. Biol. Cell 13, 2276–2288 (2002). inhibitor of 3-phosphoglycerate dehydrogenase to
13. Wieman, H. L., Wofford, J. A. & Rathmell, J. C. This study shows that AKT promotes cell survival target serine biosynthesis in cancers. Proc. Natl Acad.
Cytokine stimulation promotes glucose uptake via and growth in part by maintaining nutrient Sci. USA 113, 1778–1783 (2016).
phosphatidylinositol-3 kinase/Akt regulation of Glut1 transporter levels on the plasma membrane. 57. Yang, M. & Vousden, K. H. Serine and one-carbon
activity and trafficking. Mol. Biol. Cell 18, 1437–1446 36. Hannan, K. M. et al. mTOR-dependent regulation of metabolism in cancer. Nat. Rev. Cancer 16, 650–662
(2007). ribosomal gene transcription requires S6K1 and is (2016).
14. Rathmell, J. C., Vander Heiden, M. G., Harris, M. H., mediated by phosphorylation of the carboxy-terminal 58. Maddocks, O. D. et al. Serine starvation induces
Frauwirth, K. A. & Thompson, C. B. In the absence of activation domain of the nucleolar transcription factor stress and p53-dependent metabolic remodelling in
extrinsic signals, nutrient utilization by lymphocytes is UBF. Mol. Cell. Biol. 23, 8862–8877 (2003). cancer cells. Nature 493, 542–546 (2013).
insufficient to maintain either cell size or viability. 37. Wolfson, R. L. & Sabatini, D. M. The dawn of the age 59. Maddocks, O. D. K. et al. Modulating the therapeutic
Mol. Cell 6, 683–692 (2000). of amino acid sensors for the mTORC1 pathway. response of tumours to dietary serine and glycine
15. Fox, C. J., Hammerman, P. S. & Thompson, C. B. Cell Metab. 26, 301–309 (2017). starvation. Nature 544, 372–376 (2017).
Fuel feeds function: energy metabolism and the T cell 38. Saxton, R. A. et al. Structural basis for leucine sensing 60. Lane, A. N. & Fan, T. W. Regulation of mammalian
response. Nat. Rev. Immunol. 5, 844–852 (2005). by the Sestrin2-mTORC1 pathway. Science 351, nucleotide metabolism and biosynthesis.
16. Warburg, O., Wind, F. & Negelein, E. The metabolism 53–58 (2016). Nucleic Acids Res. 43, 2466–2485 (2015).
of tumors in the body. J. Gen. Physiol. 8, 519–530 39. Wolfson, R. L. et al. Sestrin2 is a leucine sensor for 61. Garcia-Bermudez, J. et al. Aspartate is a limiting
(1927). the mTORC1 pathway. Science 351, 43–48 (2016). metabolite for cancer cell proliferation under
17. Warburg, O. On the origin of cancer cells. Science 40. Chantranupong, L. et al. The CASTOR proteins are hypoxia and in tumours. Nat. Cell Biol. 20, 775–781
123, 309–314 (1956). arginine sensors for the mTORC1 pathway. Cell 165, (2018).
18. Kandoth, C. et al. Mutational landscape and 153–164 (2016). 62. Birsoy, K. et al. An essential role of the mitochondrial
significance across 12 major cancer types. Nature 41. Gu, X. et al. SAMTOR is an S-adenosylmethionine electron transport chain in cell proliferation is to
502, 333–339 (2013). sensor for the mTORC1 pathway. Science 358, enable aspartate synthesis. Cell 162, 540–551
19. Recondo, G., Facchinetti, F., Olaussen, K. A., Besse, B. 813–818 (2017). (2015).
& Friboulet, L. Making the first move in EGFR-driven or 42. Lu, P. D., Harding, H. P. & Ron, D. Translation 63. Sullivan, L. B. et al. Supporting aspartate biosynthesis
ALK-driven NSCLC: first-generation or next-generation reinitiation at alternative open reading frames is an essential function of respiration in proliferating
TKI? Nat. Rev. Clin. Oncol. 15, 694–708 (2018). regulates gene expression in an integrated stress cells. Cell 162, 552–563 (2015).
20. Arteaga, C. L. et al. Treatment of HER2-positive breast response. J. Cell Biol. 167, 27–33 (2004). Birsoy et al. (2015) and Sullivan et al. (2015)
cancer: current status and future perspectives. 43. Harding, H. P. et al. An integrated stress response demonstrate that a major function of the
Nat. Rev. Clin. Oncol. 9, 16–32 (2011). regulates amino acid metabolism and resistance to mitochondrial ETC is to support aspartate
21. Samuels, Y. et al. High frequency of mutations of the oxidative stress. Mol. Cell 11, 619–633 (2003). production.
PIK3CA gene in human cancers. Science 304, 554 44. Sato, H. et al. Transcriptional control of cystine/ 64. Lauren, P. et al. Mitochondrial complex III is necessary
(2004). glutamate transporter gene by amino acid for endothelial cell proliferation during angiogenesis.
22. Engelman, J. A., Luo, J. & Cantley, L. C. The evolution deprivation. Biochem. Biophys. Res. Commun. 325, Nat. Metab. 1, 158–171 (2019).
of phosphatidylinositol 3-kinases as regulators of 109–116 (2004). 65. Liu, X. et al. Regulation of mitochondrial biogenesis
growth and metabolism. Nat. Rev. Genet. 7, 606–619 45. Lewerenz, J. & Maher, P. Basal levels of eIF2alpha in erythropoiesis by mTORC1-mediated protein
(2006). phosphorylation determine cellular antioxidant status translation. Nat. Cell Biol. 19, 626–638 (2017).

Nature Reviews | Molecular Cell Biology


Reviews

66. Weinberg, S. E. et al. Mitochondrial complex III is 90. Pascual, G. et al. Targeting metastasis-initiating cells 118. Camps, J. L. et al. Fibroblast-mediated acceleration
essential for suppressive function of regulatory T cells. through the fatty acid receptor CD36. Nature 541, of human epithelial tumor growth in vivo. Proc. Natl
Nature 565, 495–499 (2019). 41–45 (2017). Acad. Sci. USA 87, 75–79 (1990).
67. Alistar, A. et al. Safety and tolerability of the 91. Zhang, M. et al. Adipocyte-derived lipids mediate 119. Olumi, A. F. et al. Carcinoma-associated fibroblasts
first-in-class agent CPI-613 in combination with melanoma progression via FATP proteins. direct tumor progression of initiated human prostatic
modified FOLFIRINOX in patients with metastatic Cancer Discov. 8, 1006–1025 (2018). epithelium. Cancer Res. 59, 5002–5011 (1999).
pancreatic cancer: a single-centre, open-label, 92. Sounni, N. E. et al. Blocking lipid synthesis 120. Curtis, M. et al. Fibroblasts mobilize tumor cell
dose-escalation, phase 1 trial. Lancet Oncol. 18, overcomes tumor regrowth and metastasis after glycogen to promote proliferation and metastasis.
770–778 (2017). antiangiogenic therapy withdrawal. Cell Metab. 20, Cell Metab. 29, 141–155 (2018).
68. Molina, J. R. et al. An inhibitor of oxidative 280–294 (2014). 121. Keith, B. & Simon, M. C. Hypoxia-inducible factors,
phosphorylation exploits cancer vulnerability. 93. Hochachka, P. W., Rupert, J. L., Goldenberg, L., stem cells, and cancer. Cell 129, 465–472 (2007).
Nat. Med. 24, 1036–1046 (2018). Gleave, M. & Kozlowski, P. Going malignant: 122. Trabold, O. et al. Lactate and oxygen constitute a
69. Sullivan, L. B. et al. Aspartate is an endogenous the hypoxia-cancer connection in the prostate. fundamental regulatory mechanism in wound healing.
metabolic limitation for tumour growth. Nat. Cell Biol. Bioessays 24, 749–757 (2002). Wound Repair Regen. 11, 504–509 (2003).
20, 782–788 (2018). 94. Ackerman, D. & Simon, M. C. Hypoxia, lipids, and 123. Kamphorst, J. J. et al. Human pancreatic cancer
70. Swanson, J. A. Shaping cups into phagosomes cancer: surviving the harsh tumor microenvironment. tumors are nutrient poor and tumor cells actively
and macropinosomes. Nat. Rev. Mol. Cell Biol. 9, Trends Cell Biol. 24, 472–478 (2014). scavenge extracellular protein. Cancer Res. 75,
639–649 (2008). 95. Zhang, Z., Dales, N. A. & Winther, M. D. Opportunities 544–553 (2015).
71. Recouvreux, M. V. & Commisso, C. Macropinocytosis: and challenges in developing stearoyl-coenzyme A 124. Pan, M. et al. Regional glutamine deficiency in
a metabolic adaptation to nutrient stress in cancer. desaturase-1 inhibitors as novel therapeutics for tumours promotes dedifferentiation through inhibition
Front. Endocrinol. 8, 261 (2017). human disease. J. Med. Chem. 57, 5039–5056 of histone demethylation. Nat. Cell Biol. 18,
72. Finicle, B. T., Jayashankar, V. & Edinger, A. L. Nutrient (2014). 1090–1101 (2016).
scavenging in cancer. Nat. Rev. Cancer 18, 619–633 96. Igal, R. A. Stearoyl CoA desaturase-1: new insights 125. Muranen, T. et al. Starved epithelial cells uptake
(2018). into a central regulator of cancer metabolism. extracellular matrix for survival. Nat. Commun. 8,
73. Bloomfield, G. & Kay, R. R. Uses and abuses of Biochim. Biophys. Acta 1861, 1865–1880 (2016). 13989 (2017).
macropinocytosis. J. Cell Sci. 129, 2697–2705 97. Kamphorst, J. J. et al. Hypoxic and Ras-transformed 126. Olivares, O. et al. Collagen-derived proline promotes
(2016). cells support growth by scavenging unsaturated fatty pancreatic ductal adenocarcinoma cell survival under
74. Haigler, H. T., McKanna, J. A. & Cohen, S. Rapid acids from lysophospholipids. Proc. Natl Acad. Sci. nutrient limited conditions. Nat. Commun. 8, 16031
stimulation of pinocytosis in human carcinoma cells USA 110, 8882–8887 (2013). (2017).
A-431 by epidermal growth factor. J. Cell Biol. 83, 98. Ackerman, D. et al. Triglycerides promote lipid 127. Katheder, N. S. & Rusten, T. E. Microenvironment
82–90 (1979). homeostasis during hypoxic stress by balancing and tumors-a nurturing relationship. Autophagy 13,
75. Palm, W., Araki, J., King, B., DeMatteo, R. G. fatty acid saturation. Cell Rep. 24, 2596–2605 1241–1243 (2017).
& Thompson, C. B. Critical role for PI3-kinase in (2018). 128. Sousa, C. M. et al. Pancreatic stellate cells support
regulating the use of proteins as an amino acid source. 99. Vriens, K. et al. Evidence for an alternative fatty acid tumour metabolism through autophagic alanine
Proc. Natl Acad. Sci. USA 114, E8628–E8636 desaturation pathway increasing cancer plasticity. secretion. Nature 536, 479–483 (2016).
(2017). Nature 566, 403–406 (2019). This work shows that autophagic degradation
76. Bar-Sagi, D. & Feramisco, J. R. Induction of 100. Nosal, J. M., Switzer, R. L. & Becker, M. A. and release of cellular constituents in pancreatic
membrane ruffling and fluid-phase pinocytosis in Overexpression, purification, and characterization stellate cells can support the growth of pancreatic
quiescent fibroblasts by ras proteins. Science 233, of recombinant human 5-phosphoribosyl-1- cancer cells.
1061–1068 (1986). pyrophosphate synthetase isozymes I and II. J. Biol. 129. Nieman, K. M. et al. Adipocytes promote ovarian
77. Commisso, C. et al. Macropinocytosis of protein is Chem. 268, 10168–10175 (1993). cancer metastasis and provide energy for rapid tumor
an amino acid supply route in Ras-transformed cells. 101. Qian, X. et al. Conversion of PRPS hexamer to growth. Nat. Med. 17, 1498–1503 (2011).
Nature 497, 633–637 (2013). monomer by AMPK-mediated phosphorylation 130. Rabinowitz, J. D. & White, E. Autophagy and
This study shows that macropinocytosis promotes inhibits nucleotide synthesis in response to energy metabolism. Science 330, 1344–1348 (2010).
the use of extracellular protein as an amino acid stress. Cancer Discov. 8, 94–107 (2018). 131. Kaur, J. & Debnath, J. Autophagy at the crossroads
source in RAS-transformed cells. 102. Graves, L. M. et al. Regulation of carbamoyl of catabolism and anabolism. Nat. Rev. Mol. Cell Biol.
78. Palm, W. et al. The Utilization of Extracellular Proteins phosphate synthetase by MAP kinase. Nature 403, 16, 461–472 (2015).
as Nutrients Is Suppressed by mTORC1. Cell 162, 328–332 (2000). 132. Wyant, G. A. et al. NUFIP1 is a ribosome receptor for
259–270 (2015). This work demonstrates that 103. Sigoillot, F. D. et al. Nuclear localization and starvation-induced ribophagy. Science 360, 751–758
mTORC1 activity suppresses the use of mitogen-activated protein kinase phosphorylation of (2018).
extracellular protein as an amino acid source the multifunctional protein CAD. J. Biol. Chem. 280, 133. Amaravadi, R., Kimmelman, A. C. & White, E. Recent
through macropinocytosis. 25611–25620 (2005). insights into the function of autophagy in cancer.
79. Garcia, D. & Shaw, R. J. AMPK: mechanisms of 104. Ben-Sahra, I., Howell, J. J., Asara, J. M. Genes Dev. 30, 1913–1930 (2016).
cellular energy sensing and restoration of metabolic & Manning, B. D. Stimulation of de novo pyrimidine 134. Karsli-Uzunbas, G. et al. Autophagy is required for
balance. Mol. Cell 66, 789–800 (2017). synthesis by growth signaling through mTOR and glucose homeostasis and lung tumor maintenance.
80. Kim, S. M. et al. PTEN deficiency and AMPK activation S6K1. Science 339, 1323–1328 (2013). Cancer Discov. 4, 914–927 (2014).
promote nutrient scavenging and anabolism in 105. Robitaille, A. M. et al. Quantitative phosphoproteomics 135. Guo, J. Y. et al. Autophagy provides metabolic
prostate cancer cells. Cancer Discov. 8, 866–883 reveal mTORC1 activates de novo pyrimidine synthesis. substrates to maintain energy charge and nucleotide
(2018). Science 339, 1320–1323 (2013). pools in Ras-driven lung cancer cells. Genes Dev. 30,
81. Nofal, M., Zhang, K., Han, S. & Rabinowitz, J. D. 106. Wang, Y. & Hekimi, S. Understanding ubiquinone. 1704–1717 (2016).
mTOR inhibition restores amino acid balance in cells Trends Cell Biol. 26, 367–378 (2016). 136. Yang, S. et al. Pancreatic cancers require autophagy
dependent on catabolism of extracellular protein. 107. White, R. M. et al. DHODH modulates transcriptional for tumor growth. Genes Dev. 25, 717–729 (2011).
Mol. Cell 67, 936–946 (2017). elongation in the neural crest and melanoma. Nature 137. Yang, A. et al. Autophagy is critical for pancreatic
82. Goldberg, I. J., Eckel, R. H. & Abumrad, N. A. 471, 518–522 (2011). tumor growth and progression in tumors with p53
Regulation of fatty acid uptake into tissues: 108. Sykes, D. B. et al. Inhibition of dihydroorotate alterations. Cancer Discov. 4, 905–913 (2014).
lipoprotein lipase- and CD36-mediated pathways. dehydrogenase overcomes differentiation blockade in 138. Yang, A. et al. Autophagy sustains pancreatic
J. Lipid Res. 50, S86–S90 (2009). acute myeloid leukemia. Cell 167, 171–186 (2016). cancer growth through both cell-autonomous and
83. Menendez, J. A. & Lupu, R. Fatty acid synthase and 109. Ducker, G. S. & Rabinowitz, J. D. One-carbon nonautonomous mechanisms. Cancer Discov. 8,
the lipogenic phenotype in cancer pathogenesis. metabolism in health and disease. Cell Metab. 25, 276–287 (2018).
Nat. Rev. Cancer 7, 763–777 (2007). 27–42 (2017). 139. Poillet-Perez, L. et al. Autophagy maintains tumour
84. Zaidi, N. et al. Lipogenesis and lipolysis: the pathways 110. Ben-Sahra, I., Hoxhaj, G., Ricoult, S. J. H., Asara, J. M. growth through circulating arginine. Nature 563,
exploited by the cancer cells to acquire fatty acids. & Manning, B. D. mTORC1 induces purine synthesis 569–573 (2018).
Prog. Lipid Res. 52, 585–589 (2013). through control of the mitochondrial tetrahydrofolate 140. Allen, E. L. et al. Differential aspartate usage identifies
85. Medes, G., Thomas, A. & Weinhouse, S. Metabolism cycle. Science 351, 728–733 (2016). a subset of cancer cells particularly dependent on
of neoplastic tissue. IV. A study of lipid synthesis in 111. Shi, Y., Evans, J. E. & Rock, K. L. Molecular identification OGDH. Cell Rep. 17, 876–890 (2016).
neoplastic tissue slices in vitro. Cancer Res. 13, of a danger signal that alerts the immune system to 141. Ilic, N. et al. PIK3CA mutant tumors depend on
27–29 (1953). dying cells. Nature 425, 516–521 (2003). oxoglutarate dehydrogenase. Proc. Natl Acad. Sci.
86. Gansler, T. S., Hardman, W. 3rd, Hunt, D. A., 112. Boeynaems, J. M. & Communi, D. Modulation of USA 114, E3434–E3443 (2017).
Schaffel, S. & Hennigar, R. A. Increased expression inflammation by extracellular nucleotides. J. Invest. 142. Ye, J. et al. Serine catabolism regulates mitochondrial
of fatty acid synthase (OA-519) in ovarian neoplasms Dermatol. 126, 943–944 (2006). redox control during hypoxia. Cancer Discov. 4,
predicts shorter survival. Hum. Pathol. 28, 686–692 113. Scheibner, K. A. et al. Hyaluronan fragments act as 1406–1417 (2014).
(1997). an endogenous danger signal by engaging TLR2. 143. Muir, A. et al. Environmental cystine drives glutamine
87. Sebastiani, V. et al. Fatty acid synthase is a marker of J. Immunol. 177, 1272–1281 (2006). anaplerosis and sensitizes cancer cells to glutaminase
increased risk of recurrence in endometrial carcinoma. 114. Singer, A. J. & Clark, R. A. Cutaneous wound healing. inhibition. eLife 6, e27713 (2017).
Gynecol. Oncol. 92, 101–105 (2004). N. Engl. J. Med. 341, 738–746 (1999). 144. Shin, C. S. et al. The glutamate/cystine xCT antiporter
88. Visca, P. et al. Fatty acid synthase (FAS) is a marker 115. Kalluri, R. The biology and function of fibroblasts in antagonizes glutamine metabolism and reduces
of increased risk of recurrence in lung carcinoma. cancer. Nat. Rev. Cancer 16, 582–598 (2016). nutrient flexibility. Nat. Commun. 8, 15074 (2017).
Anticancer Res. 24, 4169–4173 (2004). 116. Dvorak, H. F. Tumors: wounds that do not heal-redux. 145. Kong, H. & Chandel, N. S. Regulation of redox balance
89. Mashima, T., Seimiya, H. & Tsuruo, T. De novo fatty- Cancer Immunol. Res. 3, 1–11 (2015). in cancer and T cells. J. Biol. Chem. 293, 7499–7507
acid synthesis and related pathways as molecular 117. Sullivan, W. J. et al. Extracellular matrix remodeling (2018).
targets for cancer therapy. Br. J. Cancer 100, regulates glucose metabolism through TXNIP 146. Tonks, N. K. Redox redux: revisiting PTPs and the
1369–1372 (2009). destabilization. Cell 175, 117–132 (2018). control of cell signaling. Cell 121, 667–670 (2005).

www.nature.com/nrm
Reviews

147. Tormos, K. V. et al. Mitochondrial complex III ROS 153. Luengo, A., Gui, D. Y. & Vander Heiden, M. G. Author contributions
regulate adipocyte differentiation. Cell Metab. 14, Targeting metabolism for cancer therapy. Cell Chem. The authors contributed equally to all aspects of the article.
537–544 (2011). Biol. 24, 1161–1180 (2017).
148. Weinberg, F. et al. Mitochondrial metabolism and 154. Parker, W. B. Enzymology of purine and pyrimidine Competing interests
ROS generation are essential for Kras-mediated antimetabolites used in the treatment of cancer. C.B.T. is a founder of Agios Pharmaceuticals and a member
tumorigenicity. Proc. Natl Acad. Sci. USA 107, Chem. Rev. 109, 2880–2893 (2009). of its scientific advisory board. He is also a former member of
8788–8793 (2010). 155. Cantley, L. C. The phosphoinositide 3-kinase pathway. the Board of Directors and stockholder of Merck and
149. Schafer, Z. T. et al. Antioxidant and oncogene rescue Science 296, 1655–1657 (2002). Charles River Laboratories. He has patents related to cellular
of metabolic defects caused by loss of matrix 156. Manning, B. D. & Cantley, L. C. AKT/PKB signaling: metabolism.
attachment. Nature 461, 109–113 (2009). navigating downstream. Cell 129, 1261–1274
150. DeNicola, G. M. et al. Oncogene-induced Nrf2 (2007). Publisher’s note
transcription promotes ROS detoxification and 157. Manning, B. D. & Toker, A. AKT/PKB signaling: Springer Nature remains neutral with regard to jurisdictional
tumorigenesis. Nature 475, 106–109 (2011). navigating the network. Cell 169, 381–405 (2017). claims in published maps and institutional affiliations.
151. Harris, I. S. et al. Glutathione and thioredoxin 158. Vivanco, I. & Sawyers, C. L. The phosphatidylinositol
antioxidant pathways synergize to drive cancer initiation 3-Kinase AKT pathway in human cancer. Nat. Rev. Reviewer information
and progression. Cancer Cell 27, 211–222 (2015). Cancer 2, 489–501 (2002). Nature Reviews Molecular Cell Biology thanks N. Chandel,
152. Piskounova, E. et al. Oxidative stress inhibits distant D. Sabatini, and other anonymous reviewer(s), for their
metastasis by human melanoma cells. Nature 527, Acknowledgements contribution to the peer review of this work.
186–191 (2015). The authors thank members of the Thompson laboratory for
This work demonstrates that melanoma cells need critical discussions during manuscript preparation. J.Z. is sup- Supplementary information
to overcome the oxidative stress barrier in order ported by the Leukemia and Lymphoma Society postdoctoral Supplementary information is available for this paper at
to successfully metastasize. fellowship. https://doi.org/10.1038/s41580-019-0123-5.

Nature Reviews | Molecular Cell Biology

You might also like