Metabolic Changes Associated With Tumor Metastasis, Part 2: Mitochondria, Lipid and Amino Acid Metabolism
Metabolic Changes Associated With Tumor Metastasis, Part 2: Mitochondria, Lipid and Amino Acid Metabolism
Metabolic Changes Associated With Tumor Metastasis, Part 2: Mitochondria, Lipid and Amino Acid Metabolism
Abstract Metabolic alterations are a hallmark of cancer Keywords Tumor metastasis · Oxidative
controlling tumor progression and metastasis. Among the phosphorylation (OXPHOS) · Electron
various metabolic phenotypes encountered in tumors, this transport chain (ETC) · Reactive oxygen
review focuses on the contributions of mitochondria, lipid species (ROS) ·
and amino acid metabolism to the metastatic process. Tumor Tricarboxylic acid cycle (TCA cycle) · Lipogenesis ·
cells require functional mitochondria to grow, proliferate Glutaminolysis · Proline metabolism
and metastasize, but shifts in mitochondrial activities confer
pro-metastatic traits encompassing increased production of Abbreviations
mitochondrial reactive oxygen species (mtROS), enhanced aKG a-Ketoglutarate
resistance to apoptosis and the increased or de novo Aco Aconitase
production of metabolic intermediates of the TCA cycle ACLY ATP-citrate lyase
behaving as oncometabolites, including succinate, fumarate, CoA Coenzyme A
and D-2-hydroxyglutarate that control energy production, CS Citrate synthase
biosynthesis and the redox state. Lipid metabolism and the
D-2HG D-2-Hydroxyglutarate
metabolism of amino acids, such as glutamine, glutamate
EMT Epithelial-to-mesenchymal transition
and proline are also currently emerging as focal control
ETC Electron transport chain
points of cancer metastasis.
eSC Embryonic stem cell
FASN Fatty acid synthase
FH Fumarate hydratase
GDH Glutamate dehydrogenase
P. E. Porporato and V. L. Payen equally contributed to this GLS Glutaminase
manuscript. HGFR Hepatocyte growth factor receptor
HIF-1 Hypoxia-inducible factor-1
Submitted as a companion paper to ‘‘Payen VL, Porporato PE, IDH Isocitrate dehydrogenase
Baselet B, Sonveaux P. Metabolic changes associated with tumor
metastasis, part 1: Tumor pH, glycolysis and the pentose phosphate
KEAP1 Kelch-like ECH-associated protein 1
pathway.’’ KRAS Kirsten Rat Sarcoma
MCL-1 Myeloid cell leukemia-1
& Pierre Sonveaux mtROS Mitochondrial reactive oxygen species
[email protected]
mTORC1 Mammalian target of rapamycin complex 1
1
Pole of Pharmacology, Institut de Recherche Expe´rimentale NF-jB Nuclear factor-jB
et Clinique (IREC), Universite´ catholique de Louvain NRF2 Nuclear factor-like 2
(UCL), Avenue Emmanuel Mounier 52, box B1.53.09, 1200 OXPHOS Oxidative phosphorylation
Brussels, Belgium
PGC-1 Peroxisome proliferator-activated
2
Radiobiology Unit, Belgian Nuclear Research Centre, receptor c coactivator-1
SCK·CEN, 2400 Mol, Belgium
1
3
P. E. Porporato et al.
1
PHD Prolylhydroxylase http://globocan.iarc.fr.
PI3K Phosphoinositide 3-kinase
PKB/Akt Protein kinase B
ROS Reactive oxygen species
SDH Succinate dehydrogenase
SOD Superoxide dismutase
SRC2 Steroid receptor coactivator 2
SREBP Sterol regulatory element-binding protein
STAT3 Signal transducer and activator of
transcription 3
TCA (cycle) Tricarboxylic acid (cycle)
TET (enzyme) Ten-eleven translocation (enzyme)
Introduction
Mitohormesis
Metastasis + +
+ +
2H 2H 2H+
ROS
I Q
Q III Cyt C IV ETC
II ROS
Pyruvate
CoASH + NAD+
Fatty acid
dehydrogenase
Fatty acid synthase
NADH + H+ + CO 2
Acetyl-CoA
Metastasis
synthesis
H 2O Citrate
Oxaloacetate CoASH
Malate Aconitase
NAD+ dehydrogenase
Malate Isocitrate
Malate NAD+
TCA
Fumarate Isocitrate
H2O hydratase dehydrogenase
Fumarate
hydratas NADH + H+ + CO2
cycle
e
H2O
Glutamate
GTP + GDP + Pi NAD(P)+ NAD(P)H + H+
CoASH
+ H2O + NH3
Glutamate Isocitrate
dehydrogenase
NH3
α-Ketoglutarate Isocitrate
968 Glutaminase
Migration
Metastasis
H2O
NADPH NADP+
Glutamine + CO2
Fig. 1 Simplified scheme highlighting the contribution of mitochon- byproduct, mitochondrial reactive oxygen species (mtROS).
dria, lipid and amino acid metabolism to tumor metastasis. The Increased mtROS levels have been proposed to increase resistance
scheme depicts a mitochondrion where enzymes are represented in to stress (mitohormesis). Furthermore, several anaplerotic reactions
italicized blue font and their substrates in bold black. Upon entering replenish the TCA cycle. Many of these reactions promote tumor
into the mitochondria, pyruvate is broken down during the catabolic metastasis, as indicated in red. Other abbreviations: HIF-1 hypoxia-
part of the tricarboxylic acid (TCA) cycle. This produces reducing inducible factor-1, MCL-1 myeloid cell leukemia-1, NF-kB nuclear
agents that fuel the electron transport chain (ETC) to generate the factor-kB, NRF2 nuclear factor-like 2
proton motive force needed for the production of ATP and, as a
generation of the gradient, which triggers the activity of the While O2 is fully reduced to water to foster ATP syn-
ATP-synthase complex at the inner mitochondrial mem- thesis in mitochondria, a small proportion is converted into
brane. Thus, protons transferred from the intermembrane superoxide [15, 16] (Fig. 1). Superoxide is a radical anion
space to the mitochondrial matrix through ATP-synthase and a highly reactive oxygen species (ROS) acting as a
along their concentration gradient provide the energy for strong oxidant. In normal circumstances, superoxide is
ATP generation in mitochondria. detoxified by superoxide dismutases (SODs) that rapidly
convert it to H2O2. SODs are excellent catalysts that are cardiolipin at the inner mitochondrial membrane where it
rate-limited primarily by the speed of diffusion of the transfers electrons from complex III to IV of the ETC.
substrate in solution [17]. When superoxide is produced Following oxidation, this interaction is repressed and
at a supraphysiological rate and ROS rise to a critical cytochromec is released from mitochondria, thus triggering
level, they can oxidize cytochrome c and impair its apoptosis [19]. However, mitochondrial ROS (mtROS) are
binding to the inner mitochondrial membrane [18]. not only triggering
Cytochrome c is normally bound to the phospholipid
apoptosis. At non-lethal levels, they are essential for cel- acquisition of metastatic traits, with circulating tumor cells
lular adaptation to stress [20] and to hypoxia through characterized by an intermediate metabolic phenotype
activation of transcription factor hypoxia-inducible factor- compared to primary and secondary tumors (see Figure 1
1 (HIF-1) [21], as well as for the induction of autophagy in companion paper). One of the advantages conferred by
[22] and mitochondrial biogenesis by activation of the an elevation of OXPHOS activity could be a stronger
transcriptional coactivator peroxisome proliferator-acti- resistance to apoptosis through the induction of a pro-ox-
vated receptor c coactivator-1 (PGC-1) [23]. idant state [38]. Altered mitochondrial metabolism has also
The initial observation by Warburg that some tumor been proposed to promote metastasis by inducing stress
cells perform glycolysis at a high rate even in presence of tolerance during nutrient deprivation [39].
oxygen prompted him to formulate the hypothesis In strong contrast to studies advocating for a contri-
according to which mitochondria are not functional in bution of OXPHOS to cancer metastasis, a number of
tumors [3]. Except for some specific cases, this hypothesis independent authors reported that interfering with mito-
has not been con- firmed [24]. Still, the perception of the chondrial activity can promote metastasis. In particular,
role of mitochondria in tumor cells has often been reduced overexpression of oncogenes such as BAX inhibitor-1
to the provision of biosynthetic intermediates for cellular [40, 41] or loss of metastasis suppressor genes such as
proliferation [25]. But mitochondria are also emerging as KISS1 [42] induces metastasis together with a switch
essential mediators of tumorigenesis. In fact, complete from an oxidative to a glycolytic metabolism. Mito-
abolition of ETC activity was shown to prevent chondrial DNA (mtDNA) mutations can also promote
tumorigenesis in a Kirsten Rat Sar- coma (KRAS)-driven metastatic dissemination from prostate to bones and the
mouse model of lung cancer [26], in murine breast cancer invasive growth of melanoma cells in vivo [43, 44].
and melanoma models [27], and it inhibited the in vivo Accordingly, specific mtDNA mutations are a marker of
growth of human breast cancer cells [28]. In models of poor prognosis in breast cancer and melanoma patients
pancreatic cancer, mitochondrial targeting of STAT3 [45, 46]. As a final example, mitochondrial stress result-
stimulated ETC activity and tumor transformation [11, 12], ing from mtDNA depletion or treatment with ionophores
further emphasizing the critical role of mitochon- dria in that uncouple ETC activity from ATP generation pro-
cancer. Along the same lines, cancer cells responsible for moted an invasive behavior in human lung cancer cells
tumor relapse following oncogene ablation specifically and C2C12 myoblasts [47, 48].
rely on OXPHOS for survival [29]. Consistently, the It has been reported that either increased or reduced
antidiabetic drug metformin, a dual AMPK activator and OXPHOS activity promotes metastatic dissemination. A
ETC Complex I inhibitor [30], acts synergistically with potential explanation of this conundrum is that two oppo-
chemotherapy to eradicate resistant tumor cells and site metabolic phenotypes might contribute to metastasis
promote cancer remission [31]. This observation has been by regulating different pathways. Indeed, on one hand,
recently linked to the activity of metformin as an inhibitor increased mitochondrial metabolism has been shown to
of ETC Complex I [30, 32]. promote resistance to apoptosis [35, 38, 49–51], whereas,
With regard to the specific contribution of OXPHOS to on the other hand, mitochondrial dysfunction can favor a
tumor metastasis, several groups challenged the view that pro-metastatic behavior either by promoting glycolytic
ETC is dispensable for tumor migration and metastasis [27, compensation [52] or by impacting the NAD?/NADH
33–37]. Disseminating cancer cells actually display redox ratio that regulates sirtuin activity, thereby directly
increased levels of mitochondrial respiration, at least in promoting tumor metastasis [53]. We recently proposed a
breast and melanoma models [35]. This is driven by the parallel but not mutually exclusive interpretation. In dif-
upregulation of PGC-1a, a master gene regulating mito- ferent tumor cell models, we indeed observed that either an
chondrial biogenesis and metabolism, making PGC-1a increased or a dysfunctional mitochondrial activity are
expression a marker of poor prognosis in invasive ductal equally capable of promoting an invasive tumor phenotype
breast carcinoma [35]. Another piece of evidence arguing [33]. We found that metabolic states of tumor cells corre-
for an important contribution of mitochondria to tumor sponding to increased TCA cycle activity or experimentally
metastasis derives from the work of Tan et al. [27] who induced ETC bottlenecking were associated with increased
demonstrated that active mitochondria are required for mtROS generation, and mtROS were a common mediator
tumor growth in vivo and that upon experimentally of metastasis for these two different metabolic phenotypes:
induced loss of mitochondrial DNA (q0 cells), cancer cells targeting mtROS with mitochon- dria-targeted antioxidants
are capable of restoring mitochondrial respiration by mitoTEMPO or mitoQ inhibited metastatic dissemination
‘‘steal- ing’’ mitochondria from host cells. Interestingly, this in a mouse melanoma B16F10 model and abolished
work further showed that tumor cells display an increased spontaneous metastatic dissemination in a model of MDA-
mitochondrial activity (elevated O2 consumption and MB-231 human breast cancer in mice
increased presence of cristae) along with the progressive
[33]. Despite the fact that mtROS can promote apoptosis, rarely progress [72]. Oncocytomas normally occur in
depending on their production level, they also behave as endocrine and exocrine tissues (e.g., thyroid, parathyroid,
second messengers for retrograde mitochondrial signaling kidneys, salivary and pituitary glands), and are character-
to the nucleus [54] (Fig. 1). Because mtROS are short-lived ized by mitochondrial hyperplasia seen as a compensatory
and compartmentalized in mitochondria [20], they can mechanism caused by complete inactivation of Complex I
indeed spatially and temporally coordinate a localized activity [72–75]. Further evidence is required to define the
signaling cascade by oxidizing specific amino acid residues overall levels of mtROS in these tumors and their gly-
[55]. One recent example comes from studies in C. elegans colytic rate. Overall, metabolic analyses of oncocytomas
where mtROS promoted cellular adaptation to stress by point at the necessity to maintain residual ETC activity for
stimulating, e.g., the p38-nuclear factor-like 2 (NRF2) tumor invasiveness.
redox-sensitive pathway [56]. This in turn primed the
antioxidant machinery, resulting in increased lifespan of the
nematodes, a phenomenon that has been defined as TCA cycle
mitohormesis [56, 57]. In tumor cells, we found that
mtROS can cause metastasis by activating the proto- The TCA cycle comprises eight consecutive reaction steps,
oncogene Src and the focal adhesion kinase Pyk2, collec- starting from the condensation of acetyl-CoA with a
tively resulting in resistance to anoikis and increased molecule of oxaloacetate by citrate synthase (CS) to form
migration, invasion, metastasis take and spontaneous citrate (Fig. 1). Interestingly, CS expression was found to
metastasis [33, 58]. Consequently, targeting mtROS gen- be increased in pancreatic ductal adenocarcinoma [76] and
eration by complete inhibition of Complex I activity or in metastatic versus benign ovarian carcinoma [77]. Evi-
using specific mitochondria-targeted antioxidants was suf- dence is controversial for its role in metastasis, as it has
ficient to abolish metastasis formation in vivo. The fact that been shown that CS downregulation either decreases (as
mtROS are relevant for metastasis has been broadly seen after transient siRNA knock-down) [77] or increases
demonstrated, from the initial reports indicating that (following stable shRNA expression) [78] invasion. One
metastatic cells accumulate more ROS than untransformed possible explanation for these opposite observations might
cells [59] to the seminal work of Ishikawa et al. [60] be an adaptive phenotype following long-term knock-down
demonstrating that specific ROS-inducing mtDNA muta- of CS.
tions are sufficient to promote metastasis. In the latter The product of the CS reaction, citrate, is then converted
study, increased levels of mtROS activated the anti-apop- to isocitrate by aconitase (Aco). This reaction requires the
totic protein myeloid cell leukemia-1 (MCL-1), a member conversion of citrate into the enzyme-bound intermediate
of the Bcl-2 family promoting tumor cell aggressiveness. cis-aconitate. Aco reactivation is particularly relevant for
To date, several specific mtDNA mutations have been tumorigenesis in prostate cancer, where Aco is normally
reported to trigger tumor progression through the promo- inactivated by the high concentration of zinc ions present
tion of ROS production [61–63]. From a molecular in prostate cells [79]. In vitro, Aco activation has also been
standpoint, several downstream targets of mtROS have associated with the metastatic behavior of PC-3 M human
been identified, including FAK [64], PYK2 [58], SNAIL prostate cancer cells [80].
[65], p38 and NRF-2 [56, 66], nuclear factor-jB (NF-jB) Isocitrate is converted to aKG by IDHs, with the con-
activation mediated by c-Src oxidation [67] and HIF-1 comitant generation of NADH by IDH3 localized in
stabilization through mtROS-mediated prolylhydroxylase mitochondria and NADPH by IDH1 and IDH2 that traffic
(PHD) inactivation [21]. These mediators, in turn promote between the cytoplasm and mitochondria. aKG is not only
increased cell resistance to stress [68, 69]. Due to the a metabolic intermediate of the TCA cycle but also an
particular nature of ROS, it is important to consider mtROS important co-substrate of several enzymes, especially the
in a spatial context. Consequently, there are important vast family of oxygenases that performs functions ranging
caveats to consider when targeting mtROS with general from collagen synthesis to histone demethylation [81]. It is
antioxidants that can alter the total cellular ROS pool in therefore not surprising that aKG controls complex bio-
tumor and host cells and interfere with therapy, explaining logic functions, with, for example, high levels of aKG
the disappointing results of several clinical trials having promoting the maintenance of the pluripotency of embry-
treated cancer patients with general antioxidants [58, 70]. onic stem cells (eSCs) by regulating whole genome
Although, glycolytic compensation following mtDNA methylation [82]. One of the classes of oxygenases
mutation certainly contributes to the pro-metastatic phe- requiring aKG as a co-substrate is PHDs, i.e., prolylhy-
notype [42], it is not sufficient by itself [71]. In droxylases that behave as oxygen sensors. PHD2 in
oncocytoma, for example, total loss of Complex I activity particular tags HIF-1 subunit a for proteasome-mediated
was reported to correlate with the emergence of tumors that degradation under normoxia [83], and low levels of aKG
would therefore theoretically promote normoxic HIF-1 papillary renal cell cancers, sarcomas, pheochromocytomas
activation and tumor progression. aKG also mediates the and paragangliomas [98–100]. Mechanistically, succinate
activation of mammalian target of rapamycin complex 1 and fumarate are competitive inhibitors of aKG-dependent
(mTORC1) in synergy with leucine [84]. It increases the dioxygenases (i.e., enzymes that catalyze the incorporation
lifespan of C. elegans by promoting a moderate mito- of the two atoms of oxygen of O 2 into a substrate). They
chondrial inhibition through binding to ATP-synthase [85]. promote HIF-1 stabilization by inhibiting PHDs [97] and
Biological activities of aKG are thus numerous. Yet, induce a whole genome epigenetic dysregulation by
blocking the anaplerotic reactions supplementing aKG inhibiting both histone demethylases and the ten-eleven
(such as glutaminolysis and alanine transamination) inhi- translocation (TET) family of 5-methlycytosine hydroxy-
bits cell transformation and tumor invasion [26, 86], while lases [5, 101, 102]. Both metabolites thus share a common
supplementation with cell-permeable aKG is sufficient to way of inducing transformation [103]. In addition, fuma-
rescue the clonogenic potential of tumor cells [86]. High rate accumulation results in a spontaneous reaction leading
intracellular levels of aKG are likely pro-metastastic, but to posttranslational modification of cysteines in proteins
because aKG also promotes transformation, specific known as ‘succination’ (i.e., a chemical modification of
research is still required to discriminate between its impact proteins formed by a Michael addition reaction between
on tumor transformation and metastasis. Beyond the fact fumarate and thiol groups) [104]. Succination notably
that it catalyzes aKG production, the IDH reaction is of impairs the function of Kelch-like ECH-associated protein
particular interest because heterozygous mutations of 1 (KEAP1), thus relieving transcription factor NRF2 from
IDH1 and IDH2 are quite common in gliomas [87]. These KEAP1-mediated inhibition and promoting an antioxidant
mutations can result in a neomorphic enzymatic activity response [105, 106]. In contrast, succination can also affect
that allows heterodimeric IDH complexes to catalyze the glutathione, promoting oxidative stress [107]. Despite the
conversion of aKG into the oncometabolite D-2-hydrox- vast influence of succinate and fumarate on epigenetic
yglutarate (D-2HG) [6]. Mutations of IDH1 and IDH2 regulation and signaling pathways, only a limited amount
genes have been found in colon cancer [88], acute myeloid of evidence links these oncometabolites to metastatic
leukemia [89] and enchondroma [90]. Despite the fact that progression. While the experimental re-expression of FH in
D-2HG promotes tumorigenesis by altering DNA-methy- a FH-deficient renal cell carcinoma line impaired tumor
lation genome-wide (primarily by antagonizing aKG- cell migration and invasion in vitro [108], treatment with
dependent dioxygenases) [91], limited evidence links the dimethylfumarate, a cell-permeable form of fumarate,
IDH1 and IDH2 mutations to an increased metastatic strongly reduced invasion and metastasis formation in
potential [92]. melanoma [109–111]. To date, only the loss of SDH5 has
In the TCA cycle, aKG is then sequentially converted to been clearly shown to drive the acquisition of a mes-
succinyl-CoA (with the generation of NADH), succinate enchymal and prometastatic phenotype in lung cancer
(with the concurrent generation of GTP) and fumarate (by cells, further correlating with reduced levels of SDH5 in a
SDH) (Fig. 1). FH then converts fumarate into malate, small group of metastatic versus non-metastatic lung
which is used to generate oxaloacetate by malate dehy- patients [112]. However, no evidence links this effect to the
drogenase. This final reaction produces one additional enzymatic activity of SDH. Rather, SDH5 was found to
molecule of NADH from NAD?. SDH is a macromolecular form a complex with glycogen synthase kinase (GSK-3b, a
complex composed of four different subunits located on mediator of b-catenin degradation) and protein phos-
the internal face of the inner mitochondrial membrane. A phatase A (PP2A): when present, SDH5 activates GSK-3b
major characteristic of this enzymatic complex is that it is and prevents the epithelial-to-mesenchymal transition
shared between the TCA cycle and the ETC where it (EMT). When SDH5 is lost, b-catenin accumulates,
composes Complex II. Indeed, following succinate oxida- translocates to the cell nucleus and promotes EMT [112].
tion, SDH covalently binds a molecule of FAD that is The TCA cycle is amphibolic: it not only mediates
reduced to FADH2, which is followed by the transfer of catabolic reactions aimed at energy production but also
two electrons from FADH2 to ubiquinone (also known as produces precursors for cell growth. Cancer cells are cap-
coenzyme Q [CoQ]), yielding ubiquinol [CoQH2] in the able of reducing aKG to isocitrate, which has been termed
ETC. Mutations of SDH5, the protein responsible for reductive or reverse TCA cycle [113], ultimately increasing
covalently attaching FAD to SDH, are causally linked to citrate and acetyl-CoA levels to promote lipogenesis.
tumorigenesis [7, 8, 93–96] as they lead to accumulation of Glutamine directly fuels this pathway, especially when
succinate, which has been identified as an oncometabolite oxygen is limiting or when mitochondria are dysfunctional
promoting transformation [97]. On the other hand, fuma- [114, 115]. The trigger for such ‘‘anti-clockwise’’ TCA
rate accumulation linked to FH deficiency promotes the cycle has been suggested to be an increased aKG/citrate
formation of hereditary uterine fibroids, skin leiomyomas, ratio [116]. Although this phenotype has been strongly
linked to tumor growth [114, 115, 117–119], little is known overexpression was sufficient to induce resistance to
about the specific contribution of reductive carboxylation apoptosis and the transformation of normal prostate cells in
to tumor metastasis. It has nevertheless been reported that mice [130]. Similarly, ACLY is required for tumor growth
steroid receptor coactivator 2 (SRC2) promotes lipogenesis and its inhibition is a promising strategy for tumor therapy
by stimulating glutamine utilization and reductive car- [131, 132].
boxylation in prostate cancer, and SRC2 is particularly Lipid synthesis is essential for the production of mem-
enriched in metastatic lesions in patients with prostate branes necessary for cell proliferation. In addition to
cancer [120, 121]. In animal models, SRC2 depletion promoting cell proliferation, data indicate that lipid synthe-
strongly reduced tumor cell viability, tumor growth and sis further contributes to tumor progression and metastasis,
spontaneous metastasis. Although, these data suggest that with a strong increase in FASN and ACLY expression in
the metabolic phenotype characterized by a pronounced breast cancer [132, 133], retinoblastoma [134], lung cancer
reductive carboxylation might promote the emergence of [132] and colon cancer [135]. Inhibition of FASN and
aggressive tumor cell clones prone to metastasis, further ACLY was found to impair the metastatic progression of
experiments are required to test this possibility. colon cancer cells by reducing CD44- and hepatocyte
growth factor receptor (HGFR)-mediated signaling, result-
ing in reduced tumor cell migration and clonogenicity on
Lipid metabolism soft agar [136]. Because lipid synthesis endows tumor cells
with an increased resistance to apoptosis [126], tumor cells
Lipid accumulation is so common in tumors that it can by can be expected to rewire their energy metabolism towards
itself be considered as a hallmark of cancer [122]. The increased lipid synthesis to overcome therapy, an
transcription factors, sterol regulatory element-binding adaptation that further confers a more aggressive
proteins -1 and -2 (SREBP-1/-2) are the main transcrip- phenotype. In con- trast, blockade of lipid synthesis can
tional regulators of the lipogenic program, inducing inhibit metastatic dissemination following anti-angiogenic
cholesterol and fatty acid biosynthesis [123, 124]. SREBPs therapy [137]. This observation holds great promise for
are downstream targets of the phosphoinositide 3-kinase future combination therapies since one of the major
(PI3K)–protein kinase B (PKB/Akt)–mTORC1 signaling drawbacks reducing the efficacy of anti-angiogenic therapy
pathway. Their inhibition represses tumor growth by is the induction of metastases [138]. One of the major
depleting lipid rafts at the plasma membrane, thereby future challenges will be the identification of the molecular
impairing Akt activation [125]. Conversely, upregulation mechanisms linking lipid synthesis to the acquisition of
of SREBP expression renders tumor cells more resistant to invasive traits.
apoptosis and makes them more aggressive, especially in Besides the role of lipid synthesis in promoting invasion
conditions where nutrient and oxygen availability are and metastasis, it is becoming clear that fatty acid oxida-
limited [126]. As a matter of fact, the SREBP signature is a tion by itself can also promote metastasis. Indeed, simple
marker of poor prognosis in glioblastoma [126]. Notably, depletion of exogenous lipids is sufficient to impair breast
the mucin 1 oncoprotein (which is overexpressed in breast cancer cell migration in vitro, even in the presence of
cancer and is important for metastatic progression [127, alternative oxidative fuels glucose and glutamine [139].
128]) and SRC2 (which is overexpressed in prostate Similarly, in the non-small cell lung cancer cell line A549,
metastatic lesions [120], see above) are upstream the addition of transforming growth factor b promotes
regulators of SREBPs and, thus, of lipid metabolism. tumor invasiveness by increasing mitochondrial lipid oxi-
Despite, further studies are required to establish a cause– dation [140]. In this model, forced lipid oxidation alone
consequence relationship, regulation of lipid metabolism was sufficient to induce EMT. In agreement, cancer-asso-
and cancer metastasis may thus be intertwined. ciated adipocytes are emerging as key regulators of
Acetyl-CoA is at the crossroad between the TCA cycle metastatic formation in both breast and ovarian cancers
and lipid synthesis, with its production being critical for where they fuel metastases by providing tumor cells with
both metabolic pathways. Essential gatekeepers regulating energy substrates [141, 142].
de novo lipid synthesis are ATP-citrate lyase (ACLY) that
converts citrate to oxaloacetate and acetyl-CoA, and fatty
acid synthase (FASN), a macromolecular complex that Amino acid metabolism
catalyzes the condensation of carbon skeletons into fatty
acids. With a few exceptions, e.g., in liver and mammary Amino acid metabolism is a central part of cellular meta-
glands, FASN expression is low in normal tissues, but it bolic homeostasis. Among all natural amino acids,
increases during transformation [129]. In the prostate, glutamine is one of the most characterized for its role in
FASN demonstrated pro-oncogenic properties, as forced tumor formation and metastasis [9]. This non-essential
amino acid is also one of the most abundant amino acids
present in bodily fluids and one of the most heavily during nutrient deprivation, either by stimulating ATP
depleted amino acid in tumor tissue, indicating the high production or by inducing ROS-mediated autophagy [154].
avidity of tumors for glutamine [143]. Glutaminolysis has This pro-oxidant activity of proline has been proposed to
been proposed to be as important as glucose metabolism in promote mitohormesis [56], i.e., adaptation to stress driven
tumors [144] and is primarily induced by the Myc onco- by mitochondria (see also above). It most probably
gene [145]. The first step of glutamine metabolism is explains the observation that increased proline metabolism
mediated by glutaminases GLS1 and GLS2 that catalyze correlates with invasiveness and resistance to oxidative
the conversion of glutamine to glutamate and ammonia stress in esophageal squamous cell cancer [155]. Along the
(NH3). Glutamate can further be deaminated to aKG by same lines, Comes et al. [156] recently reported that pro-
glutamate dehydrogenase (GDH), with the concurrent line supplementation on its own was sufficient to trigger a
production of one molecule of NADH by GDH1 or genome-wide methylation remodeling and the acquisition
NADPH by GDH2. aKG can then be used in oxidative of an EMT-like phenotype by eSC, promoting eSC
TCA cycling, reductive TCA cycling or as a co-factor for metastasis in vivo.
biochemical reactions, as detailed above. Alternatively, Finally, amino acid metabolism could also promote
glutamate can also be transaminated by the glutamic- metastasis via a tumor cell-extrinsic action. For example,
pyruvic transaminase or by glutamic-oxaloacetic transam- the seminal work of Uyttenhove et al. [157] revealed that
inases (GOT1 or GOT2). GOT2 generates aspartate in the cancer cells can escape immune defenses by stimulating
mitochondria, which may then generate oxaloacetate in the the degradation of tryptophan, an amino acid essential for
cytosol after a second transamination reaction catalyzed by T-cell replication. Indoleamine 2,3-dioxygenase is the main
GOT1. Oxaloacetate is channeled to malate dehydroge- enzyme involved in tryptophan degradation and constitutes
nase, resulting in the production of pyruvate and NADPH. a marker of tumor aggressiveness in ovarian [158], thyroid
This alternative pathway of glutamate has emerged as a [159], breast [160] and skin [161] cancers. In another
critical regulator of the redox balance in pancreatic ductal example, enhanced serine biosynthesis has been associated
adenocarcinoma [146]. Because glutamate can further be with the induction of osteoclastogenesis and increased
used for glutathione synthesis, glutamine metabolism is formation of bone metastases in breast cancer [162],
important not only for energy production but also for redox although the molecular rationale behind this obser- vation
regulation in cancer. Hence, GLS inhibition impairs tumor still remains to be elucidated.
cell migration and invasion [86] and promotes apoptosis in
cells undergoing EMT as a consequence of decreased
resistance to oxidative stress [147]. Concluding remarks
Interestingly, GDH1, the enzyme controlling NAD ?-
dependent glutamate deamination, has been reported to be In eukaryotic cells, mitochondria evolved as gatekeepers
overexpressed in metastases of gallbladder [148] and not only of energy producing metabolism (cataplerosis)
murine hepatocarcinoma [149]. GDH1 is important for but also of biosynthetic metabolism (anaplerosis) and
redox homeostasis as it controls aKG production and the apoptosis. While a vast amount of experimental data point
subsequent generation of fumarate, which activates the at the key role of mitochondria in promoting tumor cell
antioxidant enzyme glutathione peroxidase 1 [150]. Thus, growth and proliferation in primary tumors, a rather
GDH1 has been shown to promote tumor progression by limited number of studies directly addressed their con-
increasing tumor cell resistance to oxidative stress. Its tribution to the metastatic process. The emerging picture
specific contribution to the metastatic process remains to is that while mitochondrial metabolism is hijacked by
be determined. tumor cells to promote cell growth, proliferation, redox
A more complex story of amino acids and their role in homeostasis and survival, mitochondria could also act as
cancer metastasis is that of proline. On one hand, proline bioenergetic sensors conferring migratory, invasive and
oxidase, which catalyzes proline degradation, has been metastatic phenotypes to cancer cells exposed to harsh
identified as a mitochondrial tumor suppressor due to its microenvironmental conditions. Together with de novo
pro-apoptotic properties coupled to increased ROS gener- produced D-2-hydroxyglutarate following specific IDH
ation [151]. Interestingly, the Myc oncogene inhibits mutations, mtROS and TCA cycle intermediates increas-
proline degradation [145], by participating in the accu- ingly produced in (pre)metastatic tumor cells, as well as
mulation of proline in growing tumors [143, 152]. Proline glutamate originating from glutaminolysis, could be
further accumulates in tumors following degradation of the viewed as molecules involved in retrograde signaling
extracellular matrix [153] and elevated proline synthesis in from mitochondria to the cell, suggesting that in certain
glutamine catabolism [145]. On the other hand, proline circumstances, mitochondria could drive cancer
degradation can turn into an important source of energy metastasis.
The origins of the changes affecting mitochondrial le Cancer (2012-186), the Belgian Federal Agency for Nuclear
metabolism in premetastatic cells, in metastatic progenitor Control (FANC-AFCN), the Louvain Foundation and the UCL Fonds
cells and tumor cells populating the metastatic lesion and Spe´ciaux de la Recherche (FSR). Pierre Sonveaux is a F.R.S.-FNRS
Research Associate, Paolo E. Porporato a F.R.S.-FNRS Postdoctoral
whether they can/must be reversed at specific steps of the Fellow and Vale´ry L. Payen a F.R.S.-FNRS PhD Fellow. Bjorn
metastatic process remain to be determined. They could be Baselet is a grantee of the Belgian Nuclear Research Center
dependent or independent from mutations affecting geno- (SCK·CEN).
mic or mitochondrial DNA. The observation that a
metabolic switch from OXPHOS to glycolysis can also
promote the metastatic phenotype (see companion paper) References
argues for the existence of temporally well-defined meta-
1. Gupta GP, Massague J (2006) Cancer metastasis: building a
bolic adaptations along the metastatic route. Alternatively, framework. Cell 127:679–695
different metabolic phenotypes could independently pro- 2. Warburg O, Wind F, Negelein E (1927) The metabolism of
mote tumor metastasis, with the caveat that cause–effect tumors in the body. J Gen Physiol 8:519–530
relationships must still be established in most instances. In 3. Warburg O (1956) On the origin of cancer cell. Science
123:309–314
our opinion, resolving these uncertainties is a task of fun- 4. Carew JS, Huang P (2002) Mitochondrial defects in cancer. Mol
damental importance not only to improve the Cancer 1:9
understanding of the metabolism of metastatic progenitor 5. Xiao M, Yang H, Xu W, Ma S, Lin H, Zhu H, Liu L, Liu Y,
Yang C, Xu Y, Zhao S, Ye D, Xiong Y, Guan KL (2012)
cells, but also to establish a strong rationale for the Inhibition of alpha-KG-dependent histone and DNA
development of antimetastatic treatment strategies. demethylases by fumarate and succinate that are accumulated in
A further degree of complexity of the metastatic process mutations of FH and SDH tumor suppressors. Genes Dev
arises from interactions between cellular populations with 26:1326–1338
6. Dang L, White DW, Gross S, Bennett BD, Bittinger MA,
different metabolic phenotypes inside a given tumor, Driggers EM, Fantin VR, Jang HG, Jin S, Keenan MC, Marks
resulting, e.g., in metabolic symbiosis [163] and com- KM, Prins RM, Ward PS, Yen KE, Liau LM, Rabinowitz JD,
mensalism [164]. For example, cancer-associated Cantley LC, Thompson CB, Vander Heiden MG, Su SM (2009)
Cancer-associated IDH1 mutations produce 2-hydroxyglutarate.
fibroblasts can fuel the oxidative metabolism of prostate
Nature 462:739–744
cancer cells [165], which drives EMT and metastatic pro- 7. Kaelin WG Jr (2009) SDH5 mutations and familial paragan-
gression [166]. In this context, there are strong indications glioma: somewhere Warburg is smiling. Cancer Cell 16:180–
that oxidative lipid metabolism, lipogenesis and amino 182
8. Baysal BE, Ferrell RE, Willett-Brozick JE, Lawrence EC,
acid metabolism play critical roles that largely remain to be Myssiorek D, Bosch A, van der Mey A, Taschner PE, Rubin-
explored from molecular and clinical standpoints. In par- stein WS, Myers EN, Richard CW III, Cornelisse CJ, Devilee P,
ticular, despite the identification of specific metabolic Devlin B (2000) Mutations in SDHD, a mitochondrial complex
reactions that promote metastasis, it will be pivotal to II gene, in hereditary paraganglioma. Science 287:848–851
9. DeBerardinis RJ, Cheng T (2010) Q’s next: the diverse func-
understand the molecular determinants driving the meta- tions of glutamine in metabolism, cell biology and cancer.
static switch. It goes without saying that a better Oncogene 29:313–324
delineation of specific changes affecting mitochondria, 10. Payen VL, Brisson L, Dewhirst MW, Sonveaux P (2015)
lipid and amino acid metabolism could ultimately translate Common responses of tumors and wounds to hypoxia. Cancer J
21:75–87
into new, original therapeutic strategies directed against 11. Gough DJ, Corlett A, Schlessinger K, Wegrzyn J, Larner AC,
cancer metastasis. Levy DE (2009) Mitochondrial STAT3 supports Ras-dependent
Conclusively, targeting tumor metastasis for therapy oncogenic transformation. Science 324:1713–1716
12. Kang R, Loux T, Tang D, Schapiro NE, Vernon P, Livesey KM,
now requires distinguishing metabolic changes that can Krasinskas A, Lotze MT, Zeh HJ III (2012) The expression of
drive tumor metastasis from those that merely result from the receptor for advanced glycation endproducts (RAGE) is
the acquisition of the metastatic phenotype, and permissive for early pancreatic neoplasia. Proc Natl Acad Sci U
determining whether sequential metabolic changes in a S A 109:7031–7036
13. Rasola A, Sciacovelli M, Chiara F, Pantic B, Brusilow WS,
given metastatic progenitor cell along its metastatic route Bernardi P (2010) Activation of mitochondrial ERK protects
and/or independent metabolic changes in several different cancer cells from death through inhibition of the permeability
metastatic progenitor cell populations account for the transition. Proc Natl Acad Sci U S A 107:726–731
14. Ogura M, Yamaki J, Homma MK, Homma Y (2012) Mito-
metastatic process.
chondrial c-Src regulates cell survival through phosphorylation
of respiratory chain components. Biochem J 447:281–289
Acknowledgments Work at the authors’ lab is supported by a Starting 15. Muller FL, Liu Y, Van RH (2004) Complex III releases super-
Grant from the European Research Council (ERC No. 243188 oxide to both sides of the inner mitochondrial membrane. J Biol
TUMETABO), Interuniversity Attraction Pole (IAP) grant #UP7-03 Chem 279:49064–49073
from the Belgian Science Policy Office (Belspo), an Action de 16. Wellen KE, Thompson CB (2010) Cellular metabolic stress:
Recherche Concerte´e from the Communaute´ Franc¸aise de Bel- considering how cells respond to nutrient excess. Mol Cell
gique (ARC 14/19-058), the Belgian Fonds National de la Recherche 40:323–332
Scientifique (F.R.S.-FNRS), the Te´le´vie, the Belgian Fondation
contre
17. Hsu JL, Hsieh Y, Tu C, O’Connor D, Nick HS, Silverman DN tricarboxylic acid cycle and glycolytic intermediates during cell
(1996) Catalytic properties of human manganese superoxide transformation and NTPs in cancer stem cells. Proc Natl Acad Sci U S
dismutase. J Biol Chem 271:17687–17691 A 111:10574–10579
18. Ott M, Robertson JD, Gogvadze V, Zhivotovsky B, Orrenius S 33. Porporato PE, Payen VL, Perez-Escuredo J, De Saedeleer CJ,
(2002) Cytochrome c release from mitochondria proceeds by a Danhier P, Copetti T, Dhup S, Tardy M, Vazeille T, Bouzin C,
two-step process. Proc Natl Acad Sci U S A 99:1259–1263 Feron O, Michiels C, Gallez B, Sonveaux P (2014) A Mito-
19. Zamzami N, Marchetti P, Castedo M, Decaudin D, Macho A, chondrial Switch Promotes Tumor Metastasis. Cell Rep 8:754–
Hirsch T, Susin SA, Petit PX, Mignotte B, Kroemer G (1995) 766
Sequential reduction of mitochondrial transmembrane potential 34. Comito G, Calvani M, Giannoni E, Bianchini F, Calorini L,
and generation of reactive oxygen species in early programmed Torre E, Migliore C, Giordano S, Chiarugi P (2011) HIF-1alpha
cell death. J Exp Med 182:367–377 stabilization by mitochondrial ROS promotes Met-dependent
20. Sena LA, Chandel NS (2012) Physiological roles of mitochon- invasive growth and vasculogenic mimicry in melanoma cells.
drial reactive oxygen species. Mol Cell 48:158–167 Free Radic Biol Med 51:893–904
21. Brunelle JK, Bell EL, Quesada NM, Vercauteren K, Tiranti V, 35. LeBleu VS, O’Connell JT, Gonzalez Herrera KN, Wikman H,
Zeviani M, Scarpulla RC, Chandel NS (2005) Oxygen sensing Pantel K, Haigis MC, de Carvalho FM, Damascena A, Domin-
requires mitochondrial ROS but not oxidative phosphorylation. gos Chinen LT, Rocha RM, Asara JM, Kalluri R (2014) PGC-
Cell Metab 1:409–414 1alpha mediates mitochondrial biogenesis and oxidative phos-
22. Dewaele M, Maes H, Agostinis P (2010) ROS-mediated phorylation in cancer cells to promote metastasis. Nat Cell Biol
mechanisms of autophagy stimulation and their relevance in 16:992–1003
cancer therapy. Autophagy 6:838–854 36. Chen EI, Hewel J, Krueger JS, Tiraby C, Weber MR, Kralli A,
23. St-Pierre J, Drori S, Uldry M, Silvaggi JM, Rhee J, Jager S, Becker K, Yates JR III, Felding-Habermann B (2007) Adapta-
Handschin C, Zheng K, Lin J, Yang W, Simon DK, Bachoo R, tion of energy metabolism in breast cancer brain metastases.
Spiegelman BM (2006) Suppression of reactive oxygen species Cancer Res 67:1472–1486
and neurodegeneration by the PGC-1 transcriptional coactiva- 37. Caneba CA, Bellance N, Yang L, Pabst L, Nagrath D (2012)
tors. Cell 127:397–408 Pyruvate uptake is increased in highly invasive ovarian cancer
24. Vander Heiden MG, Cantley LC, Thompson CB (2009) cells under anoikis conditions for anaplerosis, mitochondrial
Understanding the Warburg effect: the metabolic requirements function, and migration. Am J Physiol Endocrinol Metab
of cell proliferation. Science 324:1029–1033 303:E1036–E1052
25. DeBerardinis RJ, Lum JJ, Hatzivassiliou G, Thompson CB 38. Chen ZX, Pervaiz S (2007) Bcl-2 induces pro-oxidant state by
(2008) The biology of cancer: metabolic reprogramming fuels engaging mitochondrial respiration in tumor cells. Cell Death
cell growth and proliferation. Cell Metab 7:11–20 Differ 14:1617–1627
26. Weinberg F, Hamanaka R, Wheaton WW, Weinberg S, Joseph J, 39. Caino MC, Chae YC, Vaira V, Ferrero S, Nosotti M, Martin
Lopez M, Kalyanaraman B, Mutlu GM, Budinger GR, Chandel NM, Weeraratna A, O’Connell M, Jernigan D, Fatatis A, Lan-
NS (2010) Mitochondrial metabolism and ROS gen- eration are guino LR, Bosari S, Altieri DC (2013) Metabolic stress
essential for Kras-mediated tumorigenicity. Proc Natl Acad Sci regulates cytoskeletal dynamics and metastasis of cancer cells. J
U S A 107:8788–8793 Clin Invest 123:2907–2920
27. Tan AS, Baty JW, Dong LF, Bezawork-Geleta A, Endaya B, 40. Lee GH, Yan C, Shin SJ, Hong SC, Ahn T, Moon A, Park SJ,
Goodwin J, Bajzikova M, Kovarova J, Peterka M, Yan B, Lee YC, Yoo WH, Kim HT, Kim DS, Chae SW, Kim HR, Chae
Pesdar EA, Sobol M, Filimonenko A, Stuart S, Vondrusova M, HJ (2010) BAX inhibitor-1 enhances cancer metastasis by
Kluckova K, Sachaphibulkij K, Rohlena J, Hozak P, Truksa J, altering glucose metabolism and activating the sodium-
Eccles D, Haupt LM, Griffiths LR, Neuzil J, Berridge MV hydrogen exchanger: the alteration of mitochondrial function.
(2015) Mitochondrial Genome Acquisition Restores Respiratory Oncogene 29:2130–2141
Function and Tumorigenic Potential of Cancer Cells without 41. Park J, Kusminski CM, Chua SC, Scherer PE (2010) Leptin
Mitochondrial DNA. Cell Metab 21:81–94 receptor signaling supports cancer cell metabolism through
28. Fogal V, Richardson AD, Karmali PP, Scheffler IE, Smith JW, suppression of mitochondrial respiration in vivo. Am J Pathol
Ruoslahti E (2010) Mitochondrial p32 protein is a critical reg- 177:3133–3144
ulator of tumor metabolism via maintenance of oxidative 42. Liu W, Beck BH, Vaidya KS, Nash KT, Feeley KP, Ballinger
phosphorylation. Mol Cell Biol 30:1303–1318 SW, Pounds KM, Denning WL, Diers AR, Landar A, Dhar A,
29. Viale A, Pettazzoni P, Lyssiotis CA, Ying H, Sanchez N, Iwakuma T, Welch DR (2014) Metastasis suppressor KISS1
Marchesini M, Carugo A, Green T, Seth S, Giuliani V, Kost- seems to reverse the Warburg effect by enhancing mitochondrial
Alimova M, Muller F, Colla S, Nezi L, Genovese G, Deem AK, biogenesis. Cancer Res 74:954–963
Kapoor A, Yao W, Brunetto E, Kang Y, Yuan M, Asara JM, 43. Govindarajan B, Sligh JE, Vincent BJ, Li M, Canter JA, Nick-
Wang YA, Heffernan TP, Kimmelman AC, Wang H, Fleming oloff BJ, Rodenburg RJ, Smeitink JA, Oberley L, Zhang Y,
JB, Cantley LC, DePinho RA, Draetta GF (2014) Oncogene Slingerland J, Arnold RS, Lambeth JD, Cohen C, Hilenski L,
ablation-resistant pancreatic cancer cells depend on mitochon- Griendling K, Martinez-Diez M, Cuezva JM, Arbiser JL (2007)
drial function. Nature 514:628–632 Overexpression of Akt converts radial growth melanoma to
30. Wheaton WW, Weinberg SE, Hamanaka RB, Soberanes S, Sul- vertical growth melanoma. J Clin Invest 117:719–729
livan LB, Anso E, Glasauer A, Dufour E, Mutlu GM, Budigner 44. Arnold RS, Sun CQ, Richards JC, Grigoriev G, Coleman IM,
GS, Chandel NS (2014) Metformin inhibits mitochondrial com- Nelson PS, Hsieh CL, Lee JK, Xu Z, Rogatko A, Osunkoya AO,
plex I of cancer cells to reduce tumorigenesis. Elife 3:e02242 Zayzafoon M, Chung L, Petros JA (2009) Mitochondrial DNA
31. Hirsch HA, Iliopoulos D, Tsichlis PN, Struhl K (2009) Met- mutation stimulates prostate cancer growth in bone stromal
formin selectively targets cancer stem cells, and acts together environment. Prostate 69:1–11
with chemotherapy to block tumor growth and prolong remis- 45. Kuo SJ, Chen M, Ma GC, Chen ST, Chang SP, Lin WY, Chen
sion. Cancer Res 69:7507–7511 YC, Lee TH, Lin TT, Liu CS (2010) Number of somatic
32. Janzer A, German NJ, Gonzalez-Herrera KN, Asara JM, Haigis mutations in the mitochondrial D-loop region indicates poor
MC, Struhl K (2014) Metformin and phenformin deplete prognosis in breast cancer, independent of TP53 mutation.
Cancer Genet Cytogenet 201:94–101
46. Ebner S, Lang R, Mueller EE, Eder W, Oeller M, Moser A, cancerous mitochondria can inhibit tumor properties of meta- static
Koller J, Paulweber B, Mayr JA, Sperl W, Kofler B (2011) cells by suppressing oncogenic pathways. PLoS One 8:e61747
Mitochondrial haplogroups, control region polymorphisms and 63. Dasgupta S, Hoque MO, Upadhyay S, Sidransky D (2008)
malignant melanoma: a study in middle European Caucasians. Mitochondrial cytochrome B gene mutation promotes tumor
PLoS ONE 6:e27192 growth in bladder cancer. Cancer Res 68:700–706
47. Amuthan G, Biswas G, Ananadatheerthavarada HK, Vijayasar- 64. Ali MH, Mungai PT, Schumacker PT (2006) Stretch-induced
athy C, Shephard HM, Avadhani NG (2002) Mitochondrial phosphorylation of focal adhesion kinase in endothelial cells:
stress-induced calcium signaling, phenotypic changes and role of mitochondrial oxidants. Am J Physiol Lung Cell Mol
invasive behavior in human lung carcinoma A549 cells. Onco- Physiol 291:L38–L45
gene 21:7839–7849 65. Radisky DC, Levy DD, Littlepage LE, Liu H, Nelson CM, Fata
48. Amuthan G, Biswas G, Zhang SY, Klein-Szanto A, Vijayasar- JE, Leake D, Godden EL, Albertson DG, Nieto MA, Werb Z,
athy C, Avadhani NG (2001) Mitochondria-to-nucleus stress Bissell MJ (2005) Rac1b and reactive oxygen species mediate
signaling induces phenotypic changes, tumor progression and MMP-3-induced EMT and genomic instability. Nature 436:123–
cell invasion. EMBO J 20:1910–1920 127
49. Samudio I, Harmancey R, Fiegl M, Kantarjian H, Konopleva M, 66. Imhoff BR, Hansen JM (2009) Extracellular redox status regu-
Korchin B, Kaluarachchi K, Bornmann W, Duvvuri S, Taegt- lates Nrf2 activation through mitochondrial reactive oxygen
meyer H, Andreeff M (2010) Pharmacologic inhibition of fatty species. Biochem J 424:491–500
acid oxidation sensitizes human leukemia cells to apoptosis 67. Lluis JM, Buricchi F, Chiarugi P, Morales A, Fernandez-Checa
induction. J Clin Invest 120:142–156 JC (2007) Dual role of mitochondrial reactive oxygen species in
50. Girnun GD (2012) The diverse role of the PPARgamma coac- hypoxia signaling: activation of nuclear factor-{kappa}B via c-
tivator 1 family of transcriptional coactivators in cancer. Semin SRC and oxidant-dependent cell death. Cancer Res 67:7368–
Cell Dev Biol 23:381–388 7377
51. Ghosh JC, Siegelin MD, Vaira V, Faversani A, Tavecchio M, 68. DeNicola GM, Karreth FA, Humpton TJ, Gopinathan A, Wei C,
Chae YC, Lisanti S, Rampini P, Giroda M, Caino MC, Seo JH, Frese K, Mangal D, Yu KH, Yeo CJ, Calhoun ES, Scrimieri F,
Kossenkov AV, Michalek RD, Schultz DC, Bosari S, Languino Winter JM, Hruban RH, Iacobuzio-Donahue C, Kern SE, Blair
LR, Altieri DC (2015) Adaptive mitochondrial reprogramming IA, Tuveson DA (2011) Oncogene-induced Nrf2 transcription
and resistance to PI3K therapy. J Natl Cancer Inst. doi:10.1093/ pro- motes ROS detoxification and tumorigenesis. Nature
jnci/dju502 475:106–109
52. Smallbone K, Gatenby RA, Gillies RJ, Maini PK, Gavaghan DJ 69. Assefa Z, Vantieghem A, Declercq W, Vandenabeele P, Van-
(2007) Metabolic changes during carcinogenesis: potential denheede JR, Merlevede W, de WP WP, Agostinis P (1999) The
impact on invasiveness. J Theor Biol 244:703–713 activation of the c-Jun N-terminal kinase and p38 mitogen-ac-
53. Santidrian AF, Matsuno-Yagi A, Ritland M, Seo BB, LeBoeuf tivated protein kinase signaling pathways protects HeLa cells
SE, Gay LJ, Yagi T, Felding-Habermann B (2013) Mitochon- from apoptosis following photodynamic therapy with hypericin.
drial complex I activity and NAD ?/NADH balance regulate J Biol Chem 274:8788–8796
breast cancer progression. J Clin Invest 123:1068–1081 70. Chandel NS, Tuveson DA (2014) The promise and perils of
54. Singh RK, Srivastava A, Kalaiarasan P, Manvati S, Chopra R, antioxidants for cancer patients. N Engl J Med 371:177–178
Bamezai RN (2014) mtDNA germ line variation mediated ROS 71. Ishikawa K, Hashizume O, Koshikawa N, Fukuda S, Nakada K,
generates retrograde signaling and induces pro-cancerous Takenaga K, Hayashi J (2008) Enhanced glycolysis induced by
metabolic features. Sci Rep 4:6571 mtDNA mutations does not regulate metastasis. FEBS Lett
55. Chiarugi P (2005) PTPs versus PTKs: the redox side of the coin. 582:3525–3530
Free Radic Res 39:353–364 72. Gasparre G, Romeo G, Rugolo M, Porcelli AM (2011) Learning
56. Zarse K, Schmeisser S, Groth M, Priebe S, Beuster G, Kuhlow from oncocytic tumors: why choose inefficient mitochondria?
D, Guthke R, Platzer M, Kahn CR, Ristow M (2012) Impaired Biochim Biophys Acta 1807:633–642
insulin/IGF1 signaling extends life span by promoting mito- 73. Tallini G (1998) Oncocytic tumours. Virchows Arch 433:5–12
chondrial L-proline catabolism to induce a transient ROS signal. 74. Gasparre G, Hervouet E, De LE, Demont J, Pennisi LF,
Cell Metab 15:451–465 Colombel M, Mege-Lechevallier F, Scoazec JY, Bonora E,
57. Ristow M (2014) Unraveling the truth about antioxidants: Smeets R, Smeitink J, Lazar V, Lespinasse J, Giraud S, Godinot
mitohormesis explains ROS-induced health benefits. Nat Med C, Romeo G, Simonnet H (2008) Clonal expansion of mutated
20:709–711 mitochondrial DNA is associated with tumor formation and
58. Porporato PE, Sonveaux P (2015) Paving the way for a thera- complex I deficiency in the benign renal oncocytoma. Hum Mol
peutic prevention of tumor metastasis with agents targeting Genet 17:986–995
mitochondrial superoxide. Mol Cell Oncol. doi:10.4161/ 75. Mayr JA, Meierhofer D, Zimmermann F, Feichtinger R, Kogler
23723548.2014.968043 C, Ratschek M, Schmeller N, Sperl W, Kofler B (2008) Loss of
59. Szatrowski TP, Nathan CF (1991) Production of large amounts complex I due to mitochondrial DNA mutations in renal onco-
of hydrogen peroxide by human tumor cells. Cancer Res cytoma. Clin Cancer Res 14:2270–2275
51:794–798 76. Schlichtholz B, Turyn J, Goyke E, Biernacki M, Jaskiewicz K,
60. Ishikawa K, Takenaga K, Akimoto M, Koshikawa N, Yam- Sledzinski Z, Swierczynski J (2005) Enhanced citrate synthase
aguchi A, Imanishi H, Nakada K, Honma Y, Hayashi J (2008) activity in human pancreatic cancer. Pancreas 30:99–104
ROS-generating mitochondrial DNA mutations can regulate 77. Chen L, Liu T, Zhou J, Wang Y, Wang X, Di W, Zhang S (2014)
tumor cell metastasis. Science 320:661–664 Citrate synthase expression affects tumor phenotype and drug
61. Chang J, Jung HJ, Jeong SH, Kim HK, Han J, Kwon HJ (2014) resistance in human ovarian carcinoma. PLoS ONE 9:e115708
A mutation in the mitochondrial protein UQCRB promotes 78. Lin CC, Cheng TL, Tsai WH, Tsai HJ, Hu KH, Chang HC, Yeh
angiogenesis through the generation of mitochondrial reactive CW, Chen YC, Liao CC, Chang WT (2012) Loss of the respi-
oxygen species. Biochem Biophys Res Commun 455:290–297 ratory enzyme citrate synthase directly links the Warburg effect
62. Kaipparettu BA, Ma Y, Park JH, Lee TL, Zhang Y, Yotnda P, to tumor malignancy. Sci Rep 2:785
Creighton CJ, Chan WY, Wong LJ (2013) Crosstalk from non-
79. Singh KK, Desouki MM, Franklin RB, Costello LC (2006) competitive inhibitor of alpha-ketoglutarate-dependent dioxy-
Mitochondrial aconitase and citrate metabolism in malignant genases. Cancer Cell 19:17–30
and nonmalignant human prostate tissues. Mol Cancer 5:14 92. Fu Y, Zheng Y, Li K, Huang R, Zheng S, An N, Liang A (2012)
80. Mycielska ME, Broke-Smith TP, Palmer CP, Beckerman R, Mutations in isocitrate dehydrogenase 2 accelerate glioma cell
Nastos T, Erguler K, Djamgoz MB (2006) Citrate enhances migration via matrix metalloproteinase-2 and 9. Biotechnol Lett
in vitro metastatic behaviours of PC-3M human prostate cancer 34:441–446
cells: status of endogenous citrate and dependence on aconitase 93. Astuti D, Douglas F, Lennard TW, Aligianis IA, Woodward ER,
and fatty acid synthase. Int J Biochem Cell Biol 38:1766–1777 Evans DG, Eng C, Latif F, Maher ER (2001) Germline SDHD
81. Loenarz C, Schofield CJ (2008) Expanding chemical biology of mutation in familial phaeochromocytoma. Lancet 357:1181–
2-oxoglutarate oxygenases. Nat Chem Biol 4:152–156 1182
82. Carey BW, Finley LW, Cross JR, Allis CD, Thompson CB 94. Neumann HP, Bausch B, McWhinney SR, Bender BU, Gimm
(2015) Intracellular alpha-ketoglutarate maintains the pluripo- O, Franke G, Schipper J, Klisch J, Altehoefer C, Zerres K,
tency of embryonic stem cells. Nature 518:413–416 Janus- zewicz A, Eng C, Smith WM, Munk R, Manz T,
83. Berra E, Richard DE, Gothie E, Pouyssegur J (2001) HIF-1- Glaesker S, Apel TW, Treier M, Reineke M, Walz MK, Hoang-
dependent transcriptional activity is required for oxygen-medi- Vu C, Brauckhoff M, Klein-Franke A, Klose P, Schmidt H,
ated HIF-1alpha degradation. FEBS Lett 491:85–90 Maier- Woelfle M, Peczkowska M, Szmigielski C, Eng C (2002)
84. Duran RV, Oppliger W, Robitaille AM, Heiserich L, Skendaj R, Germ- line mutations in nonsyndromic pheochromocytoma. N
Gottlieb E, Hall MN (2012) Glutaminolysis activates Rag- Engl J Med 346:1459–1466
mTORC1 signaling. Mol Cell 47:349–358 95. Ricketts C, Woodward ER, Killick P, Morris MR, Astuti D, Latif
85. Chin RM, Fu X, Pai MY, Vergnes L, Hwang H, Deng G, Diep F, Maher ER (2008) Germline SDHB mutations and familial
S, Lomenick B, Meli VS, Monsalve GC, Hu E, Whelan SA, renal cell carcinoma. J Natl Cancer Inst 100:1260–1262
Wang JX, Jung G, Solis GM, Fazlollahi F, Kaweeteerawat C, 96. Janeway KA, Kim SY, Lodish M, Nose V, Rustin P, Gaal J,
Quach A, Nili M, Krall AS, Godwin HA, Chang HR, Faull KF, Dahia PL, Liegl B, Ball ER, Raygada M, Lai AH, Kelly L,
Guo F, Jiang M, Trauger SA, Saghatelian A, Braas D, Christofk Hornick JL, O’Sullivan M, de Krijger RR, Dinjens WN,
HR, Clarke CF, Teitell MA, Petrascheck M, Reue K, Jung ME, Demetri GD, Antonescu CR, Fletcher JA, Helman L, Stratakis
Frand AR, Huang J (2014) The metabolite alpha-ketoglutarate CA (2011) Defects in succinate dehydrogenase in
extends lifespan by inhibiting ATP synthase and TOR. Nature gastrointestinal stromal tumors lacking KIT and PDGFRA
510:397–401 mutations. Proc Natl Acad Sci U S A 108:314–318
86. Wang JB, Erickson JW, Fuji R, Ramachandran S, Gao P, 97. Selak MA, Armour SM, MacKenzie ED, Boulahbel H, Watson
Dinavahi R, Wilson KF, Ambrosio AL, Dias SM, Dang CV, DG, Mansfield KD, Pan Y, Simon MC, Thompson CB, Gottlieb
Cerione RA (2010) Targeting mitochondrial glutaminase activ- E (2005) Succinate links TCA cycle dysfunction to oncogenesis
ity inhibits oncogenic transformation. Cancer Cell 18:207–219 by inhibiting HIF-alpha prolyl hydroxylase. Cancer Cell 7:77–
87. Yan H, Parsons DW, Jin G, McLendon R, Rasheed BA, Yuan W, 85
Kos I, Batinic-Haberle I, Jones S, Riggins GJ, Friedman H, 98. Kiuru M, Lehtonen R, Arola J, Salovaara R, Jarvinen H, Ait-
Friedman A, Reardon D, Herndon J, Kinzler KW, Velculescu tomaki K, Sjoberg J, Visakorpi T, Knuutila S, Isola J, Delahunt
VE, Vogelstein B, Bigner DD (2009) IDH1 and IDH2 mutations B, Herva R, Launonen V, Karhu A, Aaltonen LA (2002) Few FH
in gliomas. N Engl J Med 360:765–773 mutations in sporadic counterparts of tumor types observed in
88. Ward PS, Cross JR, Lu C, Weigert O, Abel-Wahab O, Levine hereditary leiomyomatosis and renal cell cancer families.
RL, Weinstock DM, Sharp KA, Thompson CB (2012) Identifi- Cancer Res 62:4554–4557
cation of additional IDH mutations associated with 99. Tomlinson IP, Alam NA, Rowan AJ, Barclay E, Jaeger EE,
oncometabolite R(-)-2-hydroxyglutarate production. Oncogene Kelsell D, Leigh I, Gorman P, Lamlum H, Rahman S, Roylance
31:2491–2498 RR, Olpin S, Bevan S, Barker K, Hearle N, Houlston RS, Kiuru
89. Mardis ER, Ding L, Dooling DJ, Larson DE, McLellan MD, M, Lehtonen R, Karhu A, Vilkki S, Laiho P, Eklund C, Vieri-
Chen K, Koboldt DC, Fulton RS, Delehaunty KD, McGrath SD, maa O, Aittomaki K, Hietala M, Sistonen P, Paetau A,
Fulton LA, Locke DP, Magrini VJ, Abbott RM, Vickery TL, Salovaara R, Herva R, Launonen V, Aaltonen LA (2002)
Reed JS, Robinson JS, Wylie T, Smith SM, Carmichael L, Germline mutations in FH predispose to dominantly inherited
Eldred JM, Harris CC, Walker J, Peck JB, Du F, Dukes AF, uterine fibroids, skin leiomyomata and papillary renal cell
Sanderson GE, Brummett AM, Clark E, McMichael JF, Meyer cancer. Nat Genet 30:406–410
RJ, Schindler JK, Pohl CS, Wallis JW, Shi X, Lin L, Schmidt H, 100. Castro-Vega LJ, Buffet A, De Cubas AA, Cascon A, Menara M,
Tang Y, Haipek C, Wiechert ME, Ivy JV, Kalicki J, Elliott G, Khalifa E, Amar L, Azriel S, Bourdeau I, Chabre O, Curras-
Ries RE, Payton JE, Westervelt P, Tomasson MH, Watson MA, Freixes M, Franco-Vidal V, Guillaud-Bataille M, Simian C,
Baty J, Heath S, Shannon WD, Nagarajan R, Link DC, Walter Morin A, Leton R, Gomez-Grana A, Pollard PJ, Rustin P,
MJ, Graubert TA, DiPersio JF, Wilson RK, Ley TJ (2009) Robledo M, Favier J, Gimenez-Roqueplo AP (2014) Germline
Recurring mutations found by sequencing an acute myeloid mutations in FH confer predisposition to malignant pheochro-
leukemia genome. N Engl J Med 361:1058–1066 mocytomas and paragangliomas. Hum Mol Genet 23:2440–
90. Hirata M, Sasaki M, Cairns RA, Inoue S, Puviindran V, Li WY, 2446
Snow BE, Jones LD, Wei Q, Sato S, Tang YJ, Nadesan P, 101. Kaelin WG Jr (2011) Cancer and altered metabolism: potential
Rockel J, Whetstone H, Poon R, Weng A, Gross S, Straley K, importance of hypoxia-inducible factor and 2-oxoglutarate-de-
Gliser C, Xu Y, Wunder J, Mak TW, Alman BA (2015) Mutant pendent dioxygenases. Cold Spring Harb Symp Quant Biol
IDH is sufficient to initiate enchondromatosis in mice. Proc Natl 76:335–345
Acad Sci USA 112:2829–2834 102. Letouze E, Martinelli C, Loriot C, Burnichon N, Abermil N,
91. Xu W, Yang H, Liu Y, Yang Y, Wang P, Kim SH, Ito S, Yang C, Ottolenghi C, Janin M, Menara M, Nguyen AT, Benit P, Buffet
Wang P, Xiao MT, Liu LX, Jiang WQ, Liu J, Zhang JY, Wang A, Marcaillou C, Bertherat J, Amar L, Rustin P, De RA,
B, Frye S, Zhang Y, Xu YH, Lei QY, Guan KL, Zhao SM, Gimenez-Roqueplo AP, Favier J (2013) SDH mutations estab-
Xiong Y (2011) Oncometabolite 2-hydroxyglutarate is a lish a hypermethylator phenotype in paraganglioma. Cancer
Cell 23:739–752
103. King A, Selak MA, Gottlieb E (2006) Succinate dehydrogenase 117. Filipp FV, Scott DA, Ronai ZA, Osterman AL, Smith JW (2012)
and fumarate hydratase: linking mitochondrial dysfunction and Reverse TCA cycle flux through isocitrate dehydrogenases 1
cancer. Oncogene 25:4675–4682 and 2 is required for lipogenesis in hypoxic melanoma cells.
104. Nagai R, Brock JW, Blatnik M, Baatz JE, Bethard J, Walla MD, Pigment Cell Melanoma Res 25:375–383
Thorpe SR, Baynes JW, Frizzell N (2007) Succination of 118. Mullen AR, Hu Z, Shi X, Jiang L, Boroughs LK, Kovacs Z,
protein thiols during adipocyte maturation: a biomarker of Boriack R, Rakheja D, Sullivan LB, Linehan WM, Chandel NS,
mitochon- drial stress. J Biol Chem 282:34219–34228 DeBerardinis RJ (2014) Oxidation of alpha-ketoglutarate is
105. Adam J, Hatipoglu E, O’Flaherty L, Ternette N, Sahgal N, required for reductive carboxylation in cancer cells with mito-
Lockstone H, Baban D, Nye E, Stamp GW, Wolhuter K, chondrial defects. Cell Rep 7:1679–1690
Stevens M, Fischer R, Carmeliet P, Maxwell PH, Pugh CW, 119. Sun RC, Denko NC (2014) Hypoxic regulation of glutamine
Frizzell N, Soga T, Kessler BM, El-Bahrawy M, Ratcliffe PJ, metabolism through HIF1 and SIAH2 supports lipid synthesis
Pollard PJ (2011) Renal cyst formation in Fh1-deficient mice is that is necessary for tumor growth. Cell Metab 19:285–292
indepen- dent of the Hif/Phd pathway: roles for fumarate in 120. Dasgupta S, Putluri N, Long W, Zhang B, Wang J, Kaushik AK,
KEAP1 succination and Nrf2 signaling. Cancer Cell 20:524– Arnold JM, Bhowmik SK, Stashi E, Brennan CA, Rajapakshe
537 K, Coarfa C, Mitsiades N, Ittmann MM, Chinnaiyan AM,
106. Ooi A, Wong JC, Petillo D, Roossien D, Perrier-Trudova V, Sreeku- mar A, O’Malley BW (2015) Coactivator SRC-2-
Whitten D, Min BW, Tan MH, Zhang Z, Yang XJ, Zhou M, dependent metabolic reprogramming mediates prostate cancer
Gardie B, Molinie V, Richard S, Tan PH, Teh BT, Furge KA survival and metastasis. J Clin Invest 125:1174–1188
(2011) An antioxidant response phenotype shared between 121. Qin J, Lee HJ, Wu SP, Lin SC, Lanz RB, Creighton CJ,
hereditary and sporadic type 2 papillary renal cell carcinoma. DeMayo FJ, Tsai SY, Tsai MJ (2014) Androgen deprivation-
Cancer Cell 20:511–523 induced NCoA2 promotes metastatic and castration-resistant
107. Zheng L, Cardaci S, Jerby L, MacKenzie ED, Sciacovelli M, prostate cancer. J Clin Invest 124:5013–5026
Johnson TI, Gaude E, King A, Leach JD, Edrada-Ebel R, 122. Hsu PP, Sabatini DM (2008) Cancer cell metabolism: warburg
Hedley A, Morrice NA, Kalna G, Blyth K, Ruppin E, Frezza C, and beyond. Cell 134:703–707
Gottlieb E (2015) Fumarate induces redox-dependent 123. Horton JD, Goldstein JL, Brown MS (2002) SREBPs: activators
senescence by modifying glutathione metabolism. Nat Commun of the complete program of cholesterol and fatty acid synthesis
6:6001 in the liver. J Clin Invest 109:1125–1131
108. Sudarshan S, Shanmugasundaram K, Naylor SL, Lin S, Livi 124. Yamauchi Y, Furukawa K, Hamamura K, Furukawa K (2011)
CB, O’Neill CF, Parekh DJ, Yeh IT, Sun LZ, Block K (2011) Positive feedback loop between PI3 K-Akt-mTORC1 signaling
Reduced expression of fumarate hydratase in clear cell renal and the lipogenic pathway boosts Akt signaling: induction of the
cancer mediates HIF-2alpha accumulation and promotes lipogenic pathway by a melanoma antigen. Cancer Res
migration and invasion. PLoS One 6:e21037 71:4989–4997
109. Loewe R, Valero T, Kremling S, Pratscher B, Kunstfeld R, 125. Griffiths B, Lewis CA, Bensaad K, Ros S, Zhang Q, Ferber EC,
Pehamberger H, Petzelbauer P (2006) Dimethylfumarate Konisti S, Peck B, Miess H, East P, Wakelam M, Harris AL,
impairs melanoma growth and metastasis. Cancer Res Schulze A (2013) Sterol regulatory element binding protein-
66:11888–11896 dependent regulation of lipid synthesis supports cell survival
110. Yamazoe Y, Tsubaki M, Matsuoka H, Satou T, Itoh T, Kusunoki and tumor growth. Cancer Metab 1:3
T, Kidera Y, Tanimori Y, Shoji K, Nakamura H, Ogaki M, 126. Lewis CA, Brault C, Peck B, Bensaad K, Griffiths B, Mitter R,
Nishiura S, Nishida S (2009) Dimethylfumarate inhibits tumor Chakravarty P, East P, Dankworth B, Alibhai D, Harris AL,
cell invasion and metastasis by suppressing the expression and Schulze A (2015) SREBP maintains lipid biosynthesis and
activities of matrix metalloproteinases in melanoma cells. Cell viability of cancer cells under lipid- and oxygen-deprived con-
Biol Int 33:1087–1094 ditions and defines a gene signature associated with poor
111. Valero T, Steele S, Neumuller K, Bracher A, Niederleithner H, survival in glioblastoma multiforme. Oncogene. doi:10.1038/
Pehamberger H, Petzelbauer P, Loewe R (2010) Combination of onc.2014.439
dacarbazine and dimethylfumarate efficiently reduces 127. Pitroda SP, Khodarev NN, Beckett MA, Kufe DW, Weichsel-
melanoma lymph node metastasis. J Invest Dermatol baum RR (2009) MUC1-induced alterations in a lipid metabolic
130:1087–1094 gene network predict response of human breast cancers to
112. Liu J, Gao L, Zhang H, Wang D, Wang M, Zhu J, Pang C, Wang tamoxifen treatment. Proc Natl Acad Sci U S A 106:5837–5841
C (2013) Succinate dehydrogenase 5 (SDH5) regulates 128. Zhao Q, Barclay M, Hilkens J, Guo X, Barrow H, Rhodes JM,
glycogen synthase kinase 3beta-beta-catenin-mediated lung Yu LG (2010) Interaction between circulating galectin-3 and
cancer metastasis. J Biol Chem 288:29965–29973 cancer-associated MUC1 enhances tumour cell homotypic
113. Holleran AL, Briscoe DA, Fiskum G, Kelleher JK (1995) Glu- aggregation and prevents anoikis. Mol Cancer 9:154
tamine metabolism in AS-30D hepatoma cells. Evidence for its 129. Flavin R, Peluso S, Nguyen PL, Loda M (2010) Fatty acid
conversion into lipids via reductive carboxylation. Mol Cell synthase as a potential therapeutic target in cancer. Future Oncol
Biochem 152:95–101 6:551–562
114. Metallo CM, Gameiro PA, Bell EL, Mattaini KR, Yang J, Hiller 130. Migita T, Ruiz S, Fornari A, Fiorentino M, Priolo C, Zadra G,
K, Jewell CM, Johnson ZR, Irvine DJ, Guarente L, Kelleher JK, Inazuka F, Grisanzio C, Palescandolo E, Shin E, Fiore C, Xie
Vander Heiden MG, Iliopoulos O, Stephanopoulos G (2012) W, Kung AL, Febbo PG, Subramanian A, Mucci L, Ma J,
Reductive glutamine metabolism by IDH1 mediates lipogenesis Signoretti S, Stampfer M, Hahn WC, Finn S, Loda M (2009)
under hypoxia. Nature 481:380–384 Fatty acid synthase: a metabolic enzyme and candidate
115. Mullen AR, Wheaton WW, Jin ES, Chen PH, Sullivan LB, oncogene in pros- tate cancer. J Natl Cancer Inst 101:519–532
Cheng T, Yang Y, Linehan WM, Chandel NS, DeBerardinis RJ 131. Hatzivassiliou G, Zhao F, Bauer DE, Andreadis C, Shaw AN,
(2012) Reductive carboxylation supports growth in tumour cells Dhanak D, Hingorani SR, Tuveson DA, Thompson CB (2005)
with defective mitochondria. Nature 481:385–388 ATP citrate lyase inhibition can suppress tumor cell growth.
116. Fendt SM, Bell EL, Keibler MA, Olenchock BA, Mayers JR, Cancer Cell 8:311–321
Wasylenko TM, Vokes NI, Guarente L, Vander Heiden MG, 132. Migita T, Narita T, Nomura K, Miyagi E, Inazuka F, Matsuura
Stephanopoulos G (2013) Reductive glutamine metabolism is a M, Ushijima M, Mashima T, Seimiya H, Satoh Y, Okumura S,
function of the alpha-ketoglutarate to citrate ratio in cells. Nat
Commun 4:2236
Nakagawa K, Ishikawa Y (2008) ATP citrate lyase: activation and 147. Ulanet DB, Couto K, Jha A, Choe S, Wang A, Woo HK,
therapeutic implications in non-small cell lung cancer. Cancer Res Steadman M, DeLaBarre B, Gross S, Driggers E, Dorsch M,
68:8547–8554 Hurov JB (2014) Mesenchymal phenotype predisposes lung
133. Alo’ PL, Visca P, Marci A, Mangoni A, Botti C, Di TU (1996) cancer cells to impaired proliferation and redox stress in
Expression of fatty acid synthase (FAS) as a predictor of recur- response to glutaminase inhibition. PLoS ONE 9:e115144
rence in stage I breast carcinoma patients. Cancer 77:474–482 148. Wang JW, Peng SY, Li JT, Wang Y, Zhang ZP, Cheng Y, Cheng
134. Camassei FD, Cozza R, Acquaviva A, Jenkner A, Rava L, DQ, Weng WH, Wu XS, Fei XZ, Quan ZW, Li JY, Li SG, Liu
Gareri R, Donfrancesco A, Bosman C, Vadala P, Hadjistilianou YB (2009) Identification of metastasis-associated pro- teins
T, Boldrini R (2003) Expression of the lipogenic enzyme fatty involved in gallbladder carcinoma metastasis by proteomic
acid synthase (FAS) in retinoblastoma and its correlation with analysis and functional exploration of chloride intracellular
tumor aggressiveness. Invest Ophthalmol Vis Sci 44:2399–2403 channel 1. Cancer Lett 281:71–81
135. Luque-Garcia JL, Martinez-Torrecuadrada JL, Epifano C, 149. Liu S, Sun MZ, Tang JW, Wang Z, Sun C, Greenaway FT
Canamero M, Babel I, Casal JI (2010) Differential protein (2008) High-performance liquid chromatography/nano-electro-
expression on the cell surface of colorectal cancer cells associ- spray ionization tandem mass spectrometry, two-dimensional
ated to tumor metastasis. Proteomics 10:940–952 difference in-gel electrophoresis and gene microarray identifi-
136. Zaytseva YY, Rychahou PG, Gulhati P, Elliott VA, Mustain WC, cation of lymphatic metastasis-associated biomarkers. Rapid
O’Connor K, Morris AJ, Sunkara M, Weiss HL, Lee EY, Evers Commun Mass Spectrom 22:3172–3178
BM (2012) Inhibition of fatty acid synthase attenuates CD44- 150. Jin L, Li D, Alesi GN, Fan J, Kang HB, Lu Z, Boggon TJ, Jin P,
associated signaling and reduces metastasis in colorectal cancer. Yi H, Wright ER, Duong D, Seyfried NT, Egnatchik R,
Cancer Res 72:1504–1517 DeBerardinis RJ, Magliocca KR, He C, Arellano ML, Khoury
137. Sounni NE, Cimino J, Blacher S, Primac I, Truong A, Maz- HJ, Shin DM, Khuri FR, Kang S (2015) Glutamate dehydro-
zucchelli G, Paye A, Calligaris D, Debois D, De TP, Mari B, De genase 1 signals through antioxidant glutathione peroxidase 1 to
PE, Noel A (2014) Blocking lipid synthesis overcomes tumor regulate redox homeostasis and tumor growth. Cancer Cell
regrowth and metastasis after antiangiogenic therapy with- 27:257–270
drawal. Cell Metab 20:280–294 151. Liu Y, Borchert GL, Surazynski A, Hu CA, Phang JM (2006)
138. Paez-Ribes M, Allen E, Hudock J, Takeda T, Okuyama H, Proline oxidase activates both intrinsic and extrinsic pathways
Vinals F, Inoue M, Bergers G, Hanahan D, Casanovas O (2009) for apoptosis: the role of ROS/superoxides, NFAT and MEK/
Antiangiogenic therapy elicits malignant progression of tumors ERK signaling. Oncogene 25:5640–5647
to increased local invasion and distant metastasis. Cancer Cell 152. Miyagi Y, Higashiyama M, Gochi A, Akaike M, Ishikawa T,
15:220–231 Miura T, Saruki N, Bando E, Kimura H, Imamura F, Moriyama
139. Antalis CJ, Uchida A, Buhman KK, Siddiqui RA (2011) M, Ikeda I, Chiba A, Oshita F, Imaizumi A, Yamamoto H,
Migration of MDA-MB-231 breast cancer cells depends on the Miyano H, Horimoto K, Tochikubo O, Mitsushima T, Yama-
availability of exogenous lipids and cholesterol esterification. kado M, Okamoto N (2011) Plasma free amino acid profiling of
Clin Exp Metastasis 28:733–741 five types of cancer patients and its application for early
140. Jiang L, Xiao L, Sugiura H, Huang X, Ali A, Kuro O, DeBer- detection. PLoS ONE 6:e24143
ardinis RJ, Boothman DA (2015) Metabolic reprogramming 153. Steven FS, Griffin MM, Itzhaki S, Al-Habib A (1980) A trypsin-
during TGFbeta1-induced epithelial-to-mesenchymal transition. like neutral protease on Ehrlich ascites cell surfaces: its role in
Oncogene 34:3908–3916 the activation of tumour-cell zymogen of collagenase. Br J
141. Dirat B, Bochet L, Dabek M, Daviaud D, Dauvillier S, Majed Cancer 42:712–721
B, Wang YY, Meulle A, Salles B, Le GS, Garrido I, Escourrou 154. Liu W, Glunde K, Bhujwalla ZM, Raman V, Sharma A, Phang
G, Valet P, Muller C (2011) Cancer-associated adipocytes JM (2012) Proline oxidase promotes tumor cell survival in
exhibit an activated phenotype and contribute to breast cancer hypoxic tumor microenvironments. Cancer Res 72:3677–3686
invasion. Cancer Res 71:2455–2465 155. Togashi Y, Arao T, Kato H, Matsumoto K, Terashima M,
142. Nieman KM, Kenny HA, Penicka CV, Ladanyi A, Buell-Gut- Hayashi H, de Velasco MA, Fujita Y, Kimura H, Yasuda T,
brod R, Zillhardt MR, Romero IL, Carey MS, Mills GB, Shiozaki H, Nishio K (2014) Frequent amplification of
Hotamisligil GS, Yamada SD, Peter ME, Gwin K, Lengyel E ORAOV1 gene in esophageal squamous cell cancer promotes
(2011) Adipocytes promote ovarian cancer metastasis and pro- an aggressive phenotype via proline metabolism and ROS pro-
vide energy for rapid tumor growth. Nat Med 17:1498–1503 duction. Oncotarget 5:2962–2973
143. Medina MA, Marquez J, Nunez DCI (1992) Interchange of 156. Comes S, Gagliardi M, Laprano N, Fico A, Cimmino A, Pala-
amino acids between tumor and host. Biochem Med Metab Biol midessi A, De CD, De FS, Angelini C, Scita G, Patriarca EJ,
48:1–7 Matarazzo MR, Minchiotti G (2013) L-Proline induces a mes-
144. Board M, Humm S, Newsholme EA (1990) Maximum activities enchymal-like invasive program in embryonic stem cells by
of key enzymes of glycolysis, glutaminolysis, pentose remodeling H3K9 and H3K36 methylation. Stem Cell Reports
phosphate pathway and tricarboxylic acid cycle in normal, 1:307–321
neoplastic and suppressed cells. Biochem J 265:503–509 157. Uyttenhove C, Pilotte L, Theate I, Stroobant V, Colau D, Par-
145. Liu W, Le A, Hancock C, Lane AN, Dang CV, Fan TW, Phang mentier N, Boon T, Van Den Eynde BJ (2003) Evidence for a
JM (2012) Reprogramming of proline and glutamine metabo- tumoral immune resistance mechanism based on tryptophan
lism contributes to the proliferative and metabolic responses degradation by indoleamine 2,3-dioxygenase. Nat Med 9:1269–
regulated by oncogenic transcription factor c-MYC. Proc Natl 1274
Acad Sci U S A 109:8983–8988 158. Tanizaki Y, Kobayashi A, Toujima S, Shiro M, Mizoguchi M,
146. Son J, Lyssiotis CA, Ying H, Wang X, Hua S, Ligorio M, Perera Mabuchi Y, Yagi S, Minami S, Takikawa O, Ino K (2014)
RM, Ferrone CR, Mullarky E, Shyh-Chang N, Kang Y, Fleming Indoleamine 2,3-dioxygenase promotes peritoneal metastasis of
JB, Bardeesy N, Asara JM, Haigis MC, DePinho RA, Cantley ovarian cancer by inducing an immunosuppressive environment.
LC, Kimmelman AC (2013) Glutamine supports pancreatic Cancer Sci 105:966–973
cancer growth through a KRAS-regulated metabolic pathway. 159. Ryu HS, Park YS, Park HJ, Chung YR, Yom CK, Ahn SH, Park
Nature 496:101–105 YJ, Park SH, Park SY (2014) Expression of indoleamine 2,3-
dioxygenase and infiltration of FOXP3 ? regulatory T cells are BF, Kelley MJ, Gallez B, Wahl ML, Feron O, Dewhirst MW (2008)
associated with aggressive features of papillary thyroid micro- Targeting lactate-fueled respiration selectively kills hypoxic tumor cells
carcinoma. Thyroid 24:1232–1240 in mice. J Clin Invest 118:3930–3942
160. Chen JY, Li CF, Kuo CC, Tsai KK, Hou MF, Hung WC (2014) 164. Pavlides S, Whitaker-Menezes D, Castello-Cros R, Flomenberg
Cancer/stroma interplay via cyclooxygenase-2 and indoleamine N, Witkiewicz AK, Frank PG, Casimiro MC, Wang C, Fortina P,
2,3-dioxygenase promotes breast cancer progression. Breast Addya S, Pestell RG, Martinez-Outschoorn UE, Sotgia F,
Cancer Res 16:410 Lisanti MP (2009) The reverse Warburg effect: aerobic gly-
161. Speeckaert R, Vermaelen K, van GN, Autier P, Lambert J, colysis in cancer associated fibroblasts and the tumor stroma.
Haspeslagh M, van GM, Thielemans K, Neyns B, Roche N, Cell Cycle 8:3984–4001
Verbeke N, Deron P, Speeckaert M, Brochez L (2012) Indo- 165. Fiaschi T, Marini A, Giannoni E, Taddei ML, Gandellini P, De
leamine 2,3-dioxygenase, a new prognostic marker in sentinel DA, Lanciotti M, Serni S, Cirri P, Chiarugi P (2012) Reciprocal
lymph nodes of melanoma patients. Eur J Cancer 48:2004–2011 metabolic reprogramming through lactate shuttle coordinately
162. Pollari S, Kakonen SM, Edgren H, Wolf M, Kohonen P, Sara H, influences tumor-stroma interplay. Cancer Res 72:5130–5140
Guise T, Nees M, Kallioniemi O (2011) Enhanced serine pro- 166. Giannoni E, Taddei ML, Morandi A, Comito G, Calvani M,
duction by bone metastatic breast cancer cells stimulates Bianchini F, Richichi B, Raugei G, Wong N, Tang D, Chiarugi P
osteoclastogenesis. Breast Cancer Res Treat 125:421–430 (2015) Targeting stromal-induced pyruvate kinase M2 nuclear
163. Sonveaux P, Vegran F, Schroeder T, Wergin MC, Verrax J, translocation impairs oxphos and prostate cancer metastatic
Rabbani ZN, De Saedeleer CJ, Kennedy KM, Diepart C, Jordan spread. Oncotarget. PMID:26183399