Cancer Metabolism
Cancer Metabolism
Cancer Metabolism
Review Article
a
Department of Oral Function and Molecular Biology, Ohu University School of Dentistry, 31-1 Misumido,
Tomita-machi, Koriyama 963-8611, Japan
b
Department of General Clinical Medicine, Ohu University School of Dentistry, 31-1 Misumido,
Tomita-machi, Koriyama 963-8611, Japan
KEYWORDS Summary Initial studies of cancer metabolism in the early 1920s found that cancer cells were
Glycolysis; phenotypically characterized by aerobic glycolysis, in that these cells favor glucose uptake and
Warburg effect; lactate production, even in the presence of oxygen. This property, called the Warburg effect, is
Glutamine considered a hallmark of cancer. The mechanism by which these cells acquire aerobic glycolysis
metabolism; has been uncovered. Acidic extracellular fluid, secreted by cancer cells, induces a malignant
Acidic extracellular phenotype, including invasion and metastasis. Cancer cells survival depends on a critical balance
pH of redox status, which is regulated by amino acid metabolism. Glutamine is extremely impor-
tant for oxidative phosphorylation and redox regulation. Cells highly dependent on glutamine
and that cannot survive with glutamine are called glutamine-addicted cells. Metabolic repro-
gramming has been observed in cancer stem cells, which have the property of self-renewal and
are resistant to chemotherapy and radiotherapy. These findings suggest that studies of cancer
metabolism can reveal methods of preventing cancer recurrence and metastasis.
© 2017 The Authors. Published by Elsevier Ltd. This is an open access article under the CC
BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
https://doi.org/10.1016/j.jdsr.2017.08.003
1882-7616/© 2017 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
Cancer metabolism 9
Contents
1. Introduction ................................................................................................................ 9
2. Glucose metabolism and its regulation......................................................................................9
2.1. Hypoxia .............................................................................................................. 9
2.2. Histone deacetylases (HDACs)........................................................................................9
2.3. Tyrosine and serine/threonine kinases .............................................................................. 11
2.4. Oncogenes and tumor-suppressor genes.............................................................................11
2.4.1. Ras....................................................................................................11
2.4.2. c-Myc ...................................................................................................... 11
2.4.3. The never in mitosis gene A-related kinase 2 (NEK2) ....................................................... 12
2.4.4. p53 ........................................................................................................ 12
2.4.5. c-Met and ErbB2 ........................................................................................... 12
3. Acidic metabolites.........................................................................................................12
3.1. Lactate ............................................................................................................. 12
3.2. Carbon dioxide and carbonic anhydrases (CAs)......................................................................13
3.3. Ketone bodies ...................................................................................................... 13
4. Acidic pHe signaling and metastasis........................................................................................13
5. Amino acid usage in cancer ................................................................................................ 14
5.1. Glutamine .......................................................................................................... 14
5.2. Redox regulation....................................................................................................15
5.3. Activation of mTORC1 .............................................................................................. 15
6. Perspective ................................................................................................................ 16
Conflict of interest ........................................................................................................ 16
Acknowledgments ......................................................................................................... 16
References ................................................................................................................ 16
Initial studies of cancer metabolism in the early 1920s Tumor cells utilize glycolysis to supply energy, even under
showed that the cancer phenotype for glucose metabolism aerobic conditions, resulting in the conversion of pyruvate
is unique, with increased abilities to take up glucose and to lactate in the extracellular space. Hypoxia stimulates lac-
produce lactate, even under aerobic conditions [1]. This tate production in tumors by activating hypoxia-inducible
pathway, called aerobic glycolysis or the Warburg effect, transcription factor 1␣ (HIF1␣)-dependent expression of
results in extracellular fluid around tumor tissue having genes such as glucose transporter 1 (GLUT1), hexokinase
acidic pH [1,2]. Indeed, the extracellular pH (pHe ) of most 2 (HK2), pyruvate kinase (PK) M2, pyruvate dehydroge-
tumor tissues is around 6.5—6.9, and may be even lower nase kinase 1 (PDK1), enolase 1 (ENO1), and lactate
(e.g., 5.7) in some cases [3—5]. However, despite lactate dehydrogenase A (LDHA) [10—15] (Fig. 1). LDHA converts
production by tumor tissue, blood lactate level is often unaf- pyruvate to lactate and PDK1 inhibits pyruvate dehydro-
fected [6], suggesting that acidity is limited locally to the genase (PDH), which converts pyruvate to acetyl-CoA to
microenvironment around tumor tissue. produce ATP by mitochondrial oxidative phosphorylation
Accumulated evidence about cancer phenotypes has (OXPHOS) [11,16—18]. This pathway facilitates lactate pro-
indicated that all cancers have in common six biologi- duction rather than OXPHOS. Hypoxia also induces the
cal capabilities acquired during multistep development: expression of monocarboxylate transporter 4 (MCT4), which
sustained proliferative signaling, evasion of growth sup- functions as a proton-coupled transporter of lactate across
pressors, resistance to cell death, replicative immortality, cell membranes [19,20]. Thus, hypoxia enhances the War-
induction of angiogenesis, and activation of invasion and burg effect, which is responsible for high lactate secretion
metastasis [7]. Later research has revealed two additional by tumor cells.
hallmarks of cancer: reprogrammed energy metabolism and
evasion of immune-mediated destruction [8]. Recent studies 2.2. Histone deacetylases (HDACs)
have shown that metabolic reprogramming regulates cancer
stemness [9]. Thus, ‘‘cancer metabolism’’ has again become Sirtuins, which are mammalian homologs of the yeast his-
an important research topic. Here, we focus on glucose and tone deacetylase Sir2, are NAD+ -dependent HDACs and con-
glutamine metabolism. sist of seven isoforms (SIRT1—7). These enzymes are involved
10 Y. Kato et al.
Figure 1 Oncogene and tumor suppressor gene products regulate glucose and glutamine metabolism in cancer. Glycolysis is the
main source of ATP production rather than oxidative phosphorylation (OXPHOS) in tumor cells. Glucose transporters and glycolysis
metabolic enzymes are up-regulated by oncogene product c-Myc. It was believed that mutation of p53 causes loss of function. More
recently, p53’s mutation-based ‘‘gain of function’’ has been accepted: e.g., IB kinase (IKK) is inhibited by wild type p53 (wtp53) but
activated by mutant p53 (mutp53). Glucose transporter 4 (GLUT4) and phosphoglycerate mutase (PGM1) activities are also regulated
by p53 in the same way. This means reprogramming of which metabolic pathway is directed to lactate when cellular transformation
occurs. This is a significant reprogramming of metabolic pathways during carcinogenesis. Hypoxia accelerates glycolysis dependency
for energy production through activation of hypoxia-inducible transcription factor 1 (HIF1). Malate and oxaloacetate (OAA) in the
TCA cycle can be metabolized to pyruvate in cytosol. Especially, this pathway is important for metabolism of glutamine, rather than
glucose, through ␣-ketoglutarate (␣-KG) (see Fig. 6). Two isozymes of glutamine-OAA transaminase (GOT) are closely associated in
this pathway. ASCT2, neutral amino acid transporter; SIRT6, distant mammalian Sir2 homolog (sirtuin 6); NEK2, never in mitosis gene
A-related kinase 2; NF-B, nuclear factor-B; HK2, hexokinase 2, TIGAR, TP-53-induced glycolysis and apoptosis regulator; PFK1/2,
6-phosphofructo 1-kinase 1/2; AMPK, AMP-activated protein kinase; ALDA/C, aldolase A/C; TPI, triosephosphate isomerase; GAPDH,
glyceraldehyde-3-phosphate-dehydrogenase; PGK1, phosphoglycerate kinase 1; PGM1, phosphoglycerate mutase 1; ENO1, enolase
1; PKM1/M2, pyruvate kinase M1/M2; LDHA, lactate dehydrogenase A; Nrf2, NF-E2-related factor 2; PDH, pyruvate dehydrogenase;
PDK1, pyruvate dehydrogenase kinase 1; PTEN, tensin homolog on chromosome ten; PINK1, PTEN-induced putative kinase 1; SCO2,
cytochrome c oxidase assembly factor 2; GLS1/2, glutaminase 1/2; GLUD1, glutamate dehydrogenase 1; TCA cycle, tricarboxylic
acid cycle.
in resistance to cellular stress, genomic stability, energy of expression of GLUT1, 6-phosphofructo 1-kinase/fructose
metabolism, aging and tumorigenesis. SIRT6, which deacety- 1,6-biphosphatase (PFK1/FBPase1), aldolase c (ALDOC),
lates histone H3K9, is significantly associated with glucose PDK1 and LDHA, whose expression can also be up-regulated
metabolism, elevating glucose up-take through induction by HIF1 as described above [21,22] (Fig. 1).
Cancer metabolism 11
PKM1 and PKM2 are enzymes that convert phospho- Figure 2 Increase in pyruvate kinase M2 (PKM2)/PKM1 ratio
enolpyruvate to pyruvate. PKM1 is constitutively active, by phosphorylation of tyrosine residue directs to glycine pro-
whereas PKM2 can be regulated by phosphorylation. Inter- duction. (A) PKM2 activity is regulated by phosphorylation in
estingly, phosphorylation at tyrosine or serine residues contrast to constitutively active PKM1. Phosphorylation of tyro-
has been found to differentially regulate PKM2 activity sine (Tyr) residue activates it whereas that of serine (Ser)
(Fig. 2). For example, fibroblast growth factor recep- residue inhibits it. (B) When PKM2/PKM1 ratio increases, the
tor 1 (FGFR1) directly phosphorylates tyrosine residues of metabolic pathway directs to pyruvate (continues glycolysis).
PKM2, inhibiting the formation of active, tetrameric PKM2 When the ratio decreases, glycolysis is prevented and metabolic
by disrupting the binding of PKM2 cofactor fructose 1,6- direction changes to serine followed by glycine. Glycine con-
biophosphate [34]. In contrast, the pp60src kinase, which densates with ␥-glutamylcysteine for glutathione synthesis (see
increases tyrosine phosphorylation of PKM2, inactivates Fig. 6).
the latter [35,36]. Thus, tyrosine kinase phosphorylation
by growth factor signaling inhibits PKM2, resulting in the
progression of anabolic metabolism in proliferating cells
2.4. Oncogenes and tumor-suppressor genes
[34,37]. Tyrosine phosphorylation-mediated inhibition of
PKM2 has been reported to result in the accumulation of 2.4.1. Ras
3-phosphoglycerate, resulting in the accumulation of serine Ras is a small G-protein that transmits signals of growth
followed by glycine. Glycine, along with cysteine and glu- factors, such as epidermal growth factor (EGF) and hepato-
tamate, are used to produce glutathione, which neutralizes cyte growth factor (HGF), and enhances glycolysis through
the effects of reactive oxygen species (ROS), as described the induction of HIF1␣ expression. K-Ras/B-raf signal
below. increases the transcription of Nrf2, which up-regulates the
In contrast to tyrosine phosphorylation, phosphorylation PKM2/PKM1 ratio and glycolytic enzymes [42] (Figs. 1 and 2).
of serine residues on PKM2 by serine/threonine kinases Nrf2 inhibits lipogenesis but increases NADPH regeneration
such as A-Raf and protein kinase ␦ (PKC␦) induces the for- and purine biosynthesis [43].
mation and stabilization of the tetrameric active form of
PKM2 [38,39]. Pim is a serine/threonine kinase that con- 2.4.2. c-Myc
sists of three isoforms (Pim-1, Pim-2 and Pim-3). Pim-2 c-Myc is a transcription factor that up-regulates the expres-
directly phosphorylates PKM2, which stimulates glycolysis sion of nucleotide metabolic enzymes [44] and cell cycle
and reduces mitochondrial respiration [40]. In addition, Pim- regulator proteins such as E2Fs and cyclins [45], and down-
2 induces the expression of genes targeted by HIF1 through regulates cyclin-dependent kinase inhibitors such as p15,
the activation of mTOR complex 1 (mTORC1) as described p21, and p27 [45]. c-Myc is one of the ‘‘Yamanaka factors’’
below [41]. in the original protocol for production of induced pluripotent
12 Y. Kato et al.
stem cells (iPS cells); this protocol has since been modified, above, signaling pathway of c-Met is shared with that
with c-Myc replaced by non-transforming L-Myc to prevent of erbB2 (EGF receptor): e.g., Ras/Raf signaling modu-
the risk of tumor formation by iPS-derived tissue [46,47]. lates PKM2/PKM1 ratio (Fig. 2) and PI3K-AKT-mTOR signaling
c-Myc directly induces the expression of genes encod- upregulates HK2 through HIF1 and c-Myc expression (Fig. 1,
ing glycolysis-related metabolic enzymes and transporters, see also Fig. 7). c-Met expression is induced by not only
including GLUT1, PFK2/FBPase2, PKM2, PDK1, ENO1, and HIF1 [65] but also wtp53 [66]. Interestingly, mutp53R175H ,
LDHA; and, together with HIF1, stimulates the expression of a common mutant, remains inducible function for c-Met
HK2 [10,11,13—15,48] (Fig. 1). Although c-Myc can synergis- expression but other mutants cannot [67]. In addition, c-
tically stimulate HIF1-induced HK2 expression, c-Myc alone Met is tightly associated with TIGAR expression and NADPH
has little effect on the induction of HK2 [10,12,48]. production [68]. Thus, growth factor signaling such as HGF
and EGF are strongly associated with glycolysis. Although
2.4.3. The never in mitosis gene A-related kinase 2 anti-cancer drugs targeting those receptor tyrosine kinases
(NEK2) have been developed and obtained clinical trials, and some
NEK2 is a transcription factor that promotes aerobic glycol- of them were approved in head neck cancer (e.g., cetuximab
ysis by increasing the PKM2/PKM1 ratio and by enhancing and erlotinib for EGF receptor and crizotinib for HGF recep-
the expression of GLUT4, HK2, ENO1, and LDHA [49] tor) [69], clinical efficacy of those drugs seems to include
(Figs. 1 and 2). All of these genes are also targeted by c-Myc the effect on glycolysis.
and HIF1.
3. Acidic metabolites
2.4.4. p53
The transcription factor p53 is a major product of the TP53 3.1. Lactate
tumor suppressor gene. Although wild type p53 (wtp53) sup-
presses the expression of GLUT1 and GLUT4, mutant p53 The distribution of lactate in frozen sections of clini-
(mutp53) enhances their expression which is known as the cally obtained tumor tissue has been successfully visualized
gain of function [50] (Fig. 1). Similarly, mutp53 upregulates using the induced metabolic bioluminescence imaging (imBI)
phosphoglycerate mutase 1 (PGM1) whereas wtp53 inhibits technique [70,71]. These studies showed that lactate con-
it [51]. HK2 induction has only been seen for mutp53 [52]. centrations in tumor tissue vary widely, from 10—20 to over
On the other hand, wtp53 upregulates the expression of the 30 mol/g-tissue weight, corresponding approximately to
TP-53-induced glycolysis and apoptosis regulator (TIGAR), 10—20 mM and >30 mM, respectively. Moreover, assessments
which functions as PFK2 [53]. TIGAR, in turn, inhibits the of clinical biopsy samples of primary cervical and head and
production of fructose 2,6-bisphosphate, an activator of neck cancers showed that survival was significantly longer
PFK1 [54], thereby inhibiting glycolysis and directing the in patients with low than with high median lactate levels
metabolism of glucose to the pentose phosphate pathway. [72,73]. These studies also showed a positive correlation
This results in the production of NADPH, which protects cells between lactate concentration and the incidence of both
against ROS-associated apoptosis [53]. TIGAR knockdown recurrence and metastasis, suggesting that lactate not only
has been shown to radiosensitize glioma cells by inhibiting fuels tumor growth but survival and metastasis after uptake
the nuclear translocation of thioredoxin-1, a redox-sensitive into the cytoplasm through MCT1/SLC16A1.
oxidoreductase [55]. Nucleoredoxin, a thioredoxin-related Lactate is produced not only by tumor cells but by fibrob-
oxidoreductase, has been reported to inhibit PFK1 activity, lasts in tumor tissue [74] (Fig. 3). These fibroblasts are
suggesting that nucleoredoxin is a regulator of the bal- ‘‘educated’’ by tumor cells, such that their properties differ
ance between glycolysis and the pentose phosphate pathway from those of ‘‘normal’’ fibroblasts. These educated fibrob-
[56]. In mitochondria, wtp53/mutp53 induces expression of lasts are also called cancer-associated fibroblasts (CAFs).
cytochrome c oxidase assembly factor 2 (SCO2), which reg- Because tumor cells can take up lactate through MCT1, CAFs
ulates the cytochrome c oxidase complex associated with supply energy to tumor cells via lactate and stroma-derived
oxidative phosphorylation [57,58]. Regulation of redox state lactate sustains tumor progression [75,76].
by wtp53/mutp53 has also been found to induce expression Lactate also functions as a ligand that binds to G-protein-
of glutaminase 2 (GLS2), which contributes to glutathione coupled receptor 81 (GPR81/HCAR1) [77] (Fig. 3). GPR81
production [59]. Loss of wtp53 activates nuclear factor B expression is high in several tumor types and promotes the
(NF-B), thereby increasing GLUT3 expression and enhanc- malignant phenotype of breast cancers [78]. Silencing of
ing glycolysis [60]. Interestingly, insulin-dependent GLUT4 GPR81 was found to inhibit tumor growth and metasta-
expression has been observed in gastric [61] and lung [62] sis in vivo by downregulating the expression of MCT1, a
cancers. GLUT4 expression can be increased by loss of wtp53 receptor essential for lactate up-take [79]. GPR81 signal-
function [50]. Because expression of insulin receptor is ing induced angiogenesis in breast cancers by activating
higher in cancer cells than in normal cells [63,64], GLUT4 the PI3K/AKT pathway, thereby inducing the expression
is thought to be associated with tumor development and of several genes, including those encoding amphiregulin,
progression. platelet-derived growth factor-BB (PDGF-BB), urokinase
type plasminogen activator (uPA) and vascular endothelial
2.4.5. c-Met and ErbB2 growth factor (VEGF); whereas GPR81 knockdown impaired
The Met and ERBB2, which are proto-oncogene, encode cell proliferation and increased apoptosis [78]. Thus, lactate
receptor tyrosine kinases knowing as HGF receptor (c- supports survival, growth, and metastatic behavior through
Met) and EGF receptor (ErbB2), respectively. As mentioned GPR81 signaling.
Cancer metabolism 13
Figure 3 Cell to cell communication by proton and acidic 3.3. Ketone bodies
metabolites (lactate and -hydroxybutyrate). Carbonic anhy-
drase (CA) catalyzes H2 O and CO2 yielding H2 CO3 followed by H+ Ketone bodies consist of acetoacetate, -hydroxybutyrate,
and HCO3 − . CA II and CA IX are located on the cytosol and plasma and acetone, although -hydroxybutyrate is not a ketone
membrane, respectively. Intracellular H+ is secreted by vacuolar compound. Ketone bodies are abundant in the liver and are
type-ATPase (v-ATPase), Na+ /H+ exchanger 1 (NHE1). Mono- observed during diabetic ketoacidosis in children with type
carboxylate transporter (MCT) functions as the lactate/H+ or 1 diabetes mellitus [111]. Although lipolysis is increased in
-hydroxybutyrate (OHB)/H+ co-transporter. MCT1 and MCT4 adipocytes of tumor patients, due to the high consumption
are associated with their up-take and secretion, respectively. of blood glucose by tumor cells, the blood levels of ketone
Intracellular HCO3 − can be secreted by Cl− /HCO3 − exchanger, bodies from the liver are not obviously enhanced [6]. Ketone
which is not shown in this figure. Upper cell: tumor cells in nor- bodies, however, may be secreted by CAFs and utilized
moxia and sufficient nutrition due to proximity to blood vessels. by tumor cells, suggesting that ketone bodies are impor-
Lower cell: cancer-associated fibroblasts (CAFs) or tumor cells tant in the microenvironment of tumor cells [74,112,113].
in hypoxic and inadequate nutrition due to distance from blood Moreover, similar to lactate, ketone bodies function as lig-
vessels. ands of GPR41/FFAR3, GPR43/FFAR2, GPR81/HCAR1, and
GPR109a/HCAR2 [77,114] (Fig. 3).
Although lactate enhances the malignant behavior of
3.2. Carbon dioxide and carbonic anhydrases (CAs)
tumor cells, ketone bodies have the opposite clinical effect,
with a ketogenic diet prolonging the overall survival rate of
Once incorporated into cells, glucose is converted to glu-
patients with glioma [115—118]. Administration of a keto-
cose 6-phosphate, which is metabolized by the glycolytic
genic diet has been thought to reduce the consumption of
and pentose phosphate pathways; the latter, called the
glucose, as ketone bodies supply an abundant amount of
secondary pathway of glycolysis, results in the produc-
acetyl-CoA. Furthermore, -hydroxybutyrate functions as
tion of ribose 5-phosphate and NADPH [80]. This pathway
an endogenous and specific inhibitor of HDACs when incor-
results in the production of one molecule of CO2 from one
porated into its transporter, such as MCT1/SLC16A1 and
molecule of glucose 6-phosphate, whereas glycolysis of glu-
sodium-coupled MCT1 (SMCT1/SLC5A8) [119].
cose 6-phosphate does not produce CO2 . Tumors express high
A study using a mouse glioma model found that a keto-
amounts of CAs, which catalyze the reaction of CO2 with
genic diet reduced the expression of the HIF-1A and CA9
H2 O to produce H2 CO3 , which dissociates to H+ and HCO3 − .
genes and the activation of NF-B, as well as suppressing
Experiments in glycolysis-impaired mice showed that CO2
angiogenesis, invasive potential and vascular permeability
derived from the pentose phosphate pathway was a main
[120]. These findings suggested that, in contrast to lactate,
cause of extracellular acidity in tumors [81]. The intracel-
ketone bodies have anti-tumor activity.
lularly yielded H+ from the dissociation of H2 CO3 secretes
into extracellular space through a proton pump/vacuolar-
type ATPase (v-ATPase) [82—84] or an Na+ /H+ exchanger 4. Acidic pHe signaling and metastasis
[85,86], whereas the HCO3 2− is secreted through a chlo-
ride exchanger coupled with an Na+ /H+ exchanger [87,88] Hyaluronidases and cathepsins have optimal activity at
(Fig. 3). Interestingly, glucose stimulates the assembly of acidic pH, allowing their efficient digestion of extracellu-
the V0 and V1 domains of v-ATPase through PI3K, result- lar matrices in an acidic pHe microenvironment [121—123].
ing in its activation [89]. Na+ /H+ exchangers localize to the Acidic pHe also affects cellular activity through an as yet
invadopodia (invasion front) [90], resulting in the front cell incompletely identified intracellular signaling cascade. Acid
surface being more acidic than the rear [91,92]. sensing ion channel 1a (ASIC1a) is an H+ gated cation chan-
14 Y. Kato et al.
Figure 5 Cell cycle dependent glutamine metabolism. Glutamine to ␣-ketoglutarate is metabolized by different enzymes depend-
ing on cell cycle status. (A) Oncogenic molecules such as c-Myc and K-ras activate glutaminase 1 (GLS1) and glutamine-oxaloacetate
transaminase 2 (GOT2) in proliferating cells. K-ras inhibits glutamate dehydrogenase 1 (GLUD1). Thus, GLS1 and GOT2 are major
metabolic enzymes in proliferating cells. (B) Wild type p53 (wtp53) not only increase in the cyclin-dependent kinase inhibitor p21
but also glutaminase 2 (GLS2). GLUD1 is not inhibited by K-ras in quiescent cells, thereby metabolizing by GLS2 and GLUD1.
Cancer metabolism 15
5.2. Redox regulation PI3K/AKT signaling activates mTOR, resulting in cell survival
and growth [160]. mTORC1 comprises five molecules; mTOR;
Glutathione is a tripeptide consisting of cysteine, gluta- the regulatory associated protein of mTOR (RAPTOR); the
mate, and glycine. Glutathione S-transferase contributes DEP domain containing mTOR interacting protein (DEPTOR);
to drug resistance [145]. Glutathione peroxidase oxidizes the proline-rich AKT substrate of 40-kDa (PRAS40); and the
glutathione in the presence of NADPH, with the resulting mammalian lethal with SEC13 protein 8 (mLST8) [161,162].
oxidized glutathione being a substrate of the enzyme glu- PI3K/AKT/mTOR signaling also induces expression of HIF1␣
tathione reductase to neutralize H2 O2 . Thus, glutathione and c-Myc [163—166], the activities of which are associated
plays major role in scavenging ROS [146—150]. Thioredoxin with glycolysis, as described above. Aberrant amino acid
reductase is another NADPH dependent enzyme that neu- signaling promotes growth and metastasis through Rab1A-
tralizes free radicals [151]. NADPH can be supplied by the dependent activation of mTORC1 [167].
pentose phosphate pathway and by the metabolic path- Glutamine, leucine, and arginine are the most potent
way synthesizing pyruvate from malate (Fig. 6). Glutamine stimuli of mTORC1 activation, resulting in autophagy [168].
can be metabolized to malate through ␣-ketoglutarate and Glutamine up-regulates the small G-protein ADP ribosyla-
aspartate [140]. tion factor 1 (ARF1), thereby activating mTOR (Fig. 7).
Increased glutathione concentrations contribute to the Unlike glutamine, leucine and arginine stimulate the recruit-
absorption of free radicals and are associated with tumorige- ment of mTORC1 to the surface of lysosomes, with the
nesis, angiogenesis, and drug resistance [146—148,150,152]. small G-proteins RagA/B affecting kinase activation. The
Acidic pHe enhances the formation of ROS by a path- GTP-binding protein RheB increases mTOR kinase activ-
way independent of MAPKs (p38 and ERK1/2) and Src ity [162,169]. In contrast, wtp53 activates AMP-activated
16 Y. Kato et al.
[21] Zhong L, Mostoslavsky R. SIRT6: a master epige- [40] Yu Z, Zhao X, Huang L, Zhang T, Yang F, Xie L, et al. Proviral
netic gatekeeper of glucose metabolism. Transcription insertion in murine lymphomas 2 (PIM2) oncogene phospho-
2010;1(1):17—21. rylates pyruvate kinase M2 (PKM2) and promotes glycolysis in
[22] Zhong L, D’Urso A, Toiber D, Sebastian C, Henry RE, Vadysiri- cancer cells. J Biol Chem 2013;288(49):35406—16.
sack DD, et al. The histone deacetylase Sirt6 regulates glucose [41] Zhang XH, Yu HL, Wang FJ, Han YL, Yang WL. Pim-2 modulates
homeostasis via Hif1␣. Cell 2010;140(2):280—93. aerobic glycolysis and energy production during the develop-
[23] Nguyen P, Lee S, Lorang-Leins D, Trepel J, Smart DK. SIRT2 ment of colorectal tumors. Int J Med Sci 2015;12(6):487—93.
interacts with -catenin to inhibit Wnt signaling output [42] DeNicola GM, Karreth FA, Humpton TJ, Gopinathan A,
in response to radiation-induced stress. Mol Cancer Res Wei C, Frese K, et al. Oncogene-induced Nrf2 transcrip-
2014;12(9):1244—53. tion promotes ROS detoxification and tumorigenesis. Nature
[24] Takai N, Desmond JC, Kumagai T, Gui D, Said JW, Whittaker 2011;475(7354):106—9.
S, et al. Histone deacetylase inhibitors have a profound anti- [43] Hayes JD, Dinkova-Kostova AT. The Nrf2 regulatory net-
growth activity in endometrial cancer cells. Clin Cancer Res work provides an interface between redox and intermediary
2004;10(3):1141—9. metabolism. Trends Biochem Sci 2014;39(4):199—218.
[25] Gui CY, Ngo L, Xu WS, Richon VM, Marks PA. Histone deacety- [44] Liu YC, Li F, Handler J, Huang CR, Xiang Y, Neretti N, et al.
lase (HDAC) inhibitor activation of p21WAF1 involves changes Global regulation of nucleotide biosynthetic genes by c-Myc.
in promoter-associated proteins, including HDAC1. Proc Natl PLoS One 2008;3(7):e2722.
Acad Sci U S A 2004;101(5):1241—6. [45] Bretones G, Delgado MD, León J. Myc and cell cycle control.
[26] Sarfstein R, Bruchim I, Fishman A, Werner H. The mecha- Biochim Biophys Acta 2015;1849(5):506—16.
nism of action of the histone deacetylase inhibitor vorinostat [46] Takahashi K, Yamanaka S. Induction of pluripotent stem
involves interaction with the insulin-like growth factor signal- cells from mouse embryonic and adult fibroblast cultures by
ing pathway. PLoS One 2011;6(9):e24468. defined factors. Cell 2006;126(4):663—76.
[27] Liang H, He S, Yang J, Jia X, Wang P, Chen X, et al. PTEN␣, [47] Okita K, Matsumura Y, Sato Y, Okada A, Morizane A, Okamoto
a PTEN isoform translated through alternative initiation, reg- S, et al. A more efficient method to generate integration-free
ulates mitochondrial function and energy metabolism. Cell human iPS cells. Nat Methods 2011;8(5):409—12.
Metab 2014;19(5):836—48. [48] Yeung SJ, Pan J, Lee MH. Roles of p53, MYC and HIF-1 in reg-
[28] Witt O, Deubzer HE, Milde T, Oehme I. HDAC family: what are ulating glycolysis − the seventh hallmark of cancer. Cell Mol
the cancer relevant targets? Cancer Lett 2009;277(1):8—21. Life Sci 2008;65(24):3981—99.
[29] Wang J, Kim TH, Ahn MY, Lee J, Jung JH, Choi WS, et al. [49] Gu Z, Xia J, Xu H, Frech I, Tricot G, Zhan F. NEK2 promotes
Sirtinol, a class III HDAC inhibitor, induces apoptotic and aerobic glycolysis in multiple myeloma through regulating
autophagic cell death in MCF-7 human breast cancer cells. splicing of pyruvate kinase. J Hematol Oncol 2017;10(1):17.
Int J Oncol 2012;41(3):1101—9. [50] Schwartzenberg-Bar-Yoseph F, Armoni M, Karnieli E. The
[30] West AC, Johnstone RW. New and emerging HDAC inhibitors tumor suppressor p53 down-regulates glucose trans-
for cancer treatment. J Clin Invest 2014;124(1):30—9. porters GLUT1 and GLUT4 gene expression. Cancer Res
[31] Marks PA, Breslow R. Dimethyl sulfoxide to vorinostat: devel- 2004;64(7):2627—33.
opment of this histone deacetylase inhibitor as an anticancer [51] Kondoh H, Lleonart ME, Gil J, Wang J, Degan P, Peters G, et al.
drug. Nat Biotechnol 2007;25(1):84—90. Glycolytic enzymes can modulate cellular life span. Cancer
[32] Zang Y, Kirk CJ, Johnson DE. Carfilzomib and oprozomib syn- Res 2005;65(1):177—85.
ergize with histone deacetylase inhibitors in head and neck [52] Mathupala SP, Heese C, Pedersen PL. Glucose catabolism in
squamous cell carcinoma models of acquired resistance to cancer cells. The type II hexokinase promoter contains func-
proteasome inhibitors. Cancer Biol Ther 2014;15(9):1142—52. tionally active response elements for the tumor suppressor
[33] Duvic M, Dimopoulos M. The safety profile of vorinostat p53. J Biol Chem 1997;272(36):22776—80.
(suberoylanilide hydroxamic acid) in hematologic malig- [53] Bensaad K, Tsuruta A, Selak MA, Vidal MN, Nakano K, Bartrons
nancies: a review of clinical studies. Cancer Treat Rev R, et al. TIGAR, a p53-inducible regulator of glycolysis and
2016;43:58—66. apoptosis. Cell 2006;126(1):107—20.
[34] Hitosugi T, Kang S, Vander Heiden MG, Chung TW, Elf S, [54] Rider MH, Kuntz DA. Hue L Fructose 2,6-bisphosphate and its
Lythgoe K, et al. Tyrosine phosphorylation inhibits PKM2 to phosphorothioate analogue. Comparison of their hydrolysis
promote the Warburg effect and tumor growth. Sci Signal and action on glycolytic and gluconeogenic enzymes. Biochem
2009;2(97):ra73. J 1988;253(2):597—601.
[35] Presek P, Reinacher M, Eigenbrodt E. Pyruvate kinase type M2 [55] Zhang H, Gu C, Yu J, Wang Z, Yuan X, Yang L, et al. Radiosen-
is phosphorylated at tyrosine residues in cells transformed by sitization of glioma cells by TP53-induced glycolysis and
Rous sarcoma virus. FEBS Lett 1988;242(1):194—8. apoptosis regulator knockdown is dependent on thioredoxin-1
[36] Presek P, Glossmann H, Eigenbrodt E, Schoner W, Rubsamen nuclear translocation. Free Radic Biol Med 2014;69:239—48.
H, Friis RR, et al. Similarities between a phosphoprotein [56] Funato Y, Hayashi T, Irino Y, Takenawa T, Miki H. Nucleore-
(pp60src )-associated protein kinase of Rous sarcoma virus and doxin regulates glucose metabolism via phosphofructokinase
a cyclic adenosine 3 :5 -monophosphate-independent protein 1. Biochem Biophys Res Commun 2013;440(4):737—42.
kinase that phosphorylates pyruvate kinase type M2 . Cancer [57] Matoba S, Kang JG, Patino WD, Wragg A, Boehm M, Gavrilova
Res 1980;40(5):1733—41. O, et al. p53 regulates mitochondrial respiration. Science
[37] Christofk HR, Vander Heiden MG, Wu N, Asara JM, Cantley 2006;312(5780):1650—3.
LC. Pyruvate kinase M2 is a phosphotyrosine-binding protein. [58] Won KY, Lim SJ, Kim GY, Kim YW, Han SA, Song JY, et al. Reg-
Nature 2008;452(7184):181—6. ulatory role of p53 in cancer metabolism via SCO2 and TIGAR
[38] Le Mellay V, Houben R, Troppmair J, Hagemann C, Mazurek in human breast cancer. Hum Pathol 2012;43(2):221—8.
S, Frey U, et al. Regulation of glycolysis by Raf protein ser- [59] Suzuki S, Tanaka T, Poyurovsky MV, Nagano H, Mayama T,
ine/threonine kinases. Adv Enzyme Regul 2002;42:317—32. Ohkubo S, et al. Phosphate-activated glutaminase (GLS2),
[39] Mazurek S, Grimm H, Boschek CB, Vaupel P, Eigenbrodt a p53-inducible regulator of glutamine metabolism and
E. Pyruvate kinase type M2: a crossroad in the tumor reactive oxygen species. Proc Natl Acad Sci U S A
metabolome. Br J Nutr 2002;87(Suppl. 1):S23—9. 2010;107(16):7461—6.
18 Y. Kato et al.
[60] Kawauchi K, Araki K, Tobiume K, Tanaka N. p53 regulates glu- in breast cancer through angiogenic factor secretion. Onco-
cose metabolism through an IKK-NF-B pathway and inhibits target 2016;7(43):70898—911.
cell transformation. Nat Cell Biol 2008;10(5):611—8. [79] Roland CL, Arumugam T, Deng D, Liu SH, Philip B, Gomez S,
[61] Noguchi Y, Marat D, Saito A, Yoshikawa T, Doi C, Fukuzawa K, et al. Cell surface lactate receptor GPR81 is crucial for cancer
et al. Expression of facilitative glucose transporters in gastric cell survival. Cancer Res 2014;74(18):5301—10.
tumors. Hepatogastroenterology 1999;46(28):2683—9. [80] Wamelink MM, Struys EA, Jakobs C. The biochemistry,
[62] Ito T, Noguchi Y, Udaka N, Kitamura H, Satoh S. Glucose metabolism and inherited defects of the pentose phosphate
transporter expression in developing fetal lungs and lung neo- pathway: a review. J Inherit Metab Dis 2008;31(6):703—17.
plasms. Histol Histopathol 1999;14(3):895—904. [81] Helmlinger G, Sckell A, Dellian M, Forbes NS, Jain RK.
[63] Frasca F, Pandini G, Scalia P, Sciacca L, Mineo R, Costantino Acid production in glycolysis-impaired tumors provides
A, et al. Insulin receptor isoform A, a newly recognized, new insights into tumor metabolism. Clin Cancer Res
high-affinity insulin-like growth factor II receptor in fetal and 2002;8(4):1284—91.
cancer cells. Mol Cell Biol 1999;19(5):3278—88. [82] Sennoune SR, Bakunts K, Martínez GM, Chua-Tuan JL, Kebir
[64] Vella V, Pandini G, Sciacca L, Mineo R, Vigneri R, Pezzino V, Y, Attaya MN, et al. Vacuolar H+ -ATPase in human breast
et al. A novel autocrine loop involving IGF-II and the insulin cancer cells with distinct metastatic potential: distribu-
receptor isoform-A stimulates growth of thyroid cancer. J Clin tion and functional activity. Am J Physiol Cell Physiol
Endocrinol Metab 2002;87(1):245—54. 2004;286(6):C1443—52.
[65] Eckerich C, Zapf S, Fillbrandt R, Loges S, Westphal M, Lam- [83] Nishisho T, Hata K, Nakanishi M, Morita Y, Sun-Wada GH, Wada
szus K. Hypoxia can induce c-Met expression in glioma cells Y, et al. The a3 isoform vacuolar type H+ -ATPase promotes dis-
and enhance SF/HGF-induced cell migration. Int J Cancer tant metastasis in the mouse B16 melanoma cells. Mol Cancer
2007;121(2):276—83. Res 2011;9(7):845—55.
[66] Seol DW, Chen Q, Smith ML, Zarnegar R. Regulation of [84] Pérez-Sayáns M, Reboiras-López MD, Somoza-Martín JM,
the c-met proto-oncogene promoter by p53. J Biol Chem Barros-Angueira F, Diz PG, Rey JM, et al. Measurement of
1999;274(6):3565—72. ATP6V1C1 expression in brush cytology samples as a diagnos-
[67] Grugan KD, Vega ME, Wong GS, Diehl JA, Bass AJ, Wong KK, tic and prognostic marker in oral squamous cell carcinoma.
et al. A common p53 mutation (R175H) activates c-Met recep- Cancer Biol Ther 2010;9(12):1057—64.
tor tyrosine kinase to enhance tumor cell invasion. Cancer Biol [85] Cardone RA, Casavola V, Reshkin SJ. The role of disturbed pH
Ther 2013;14(9):853—9. dynamics and the Na+ /H+ exchanger in metastasis. Nat Rev
[68] Lui VW, Wong EY, Ho K, Ng PK, Lau CP, Tsui SK, et al. Cancer 2005;5(10):786—95.
Inhibition of c-Met downregulates TIGAR expression and [86] Amith SR, Wilkinson JM, Fliegel L. Na+ /H+ exchanger NHE1
reduces NADPH production leading to cell death. Oncogene regulation modulates metastatic potential and epithelial-
2011;30(9):1127—34. mesenchymal transition of triple-negative breast cancer
[69] Elicin O, Ozsahin M. The latest prospects of investigational cells. Oncotarget 2016;7(16):21091—2113.
drugs for head and neck cancer. Expert Opin Investig Drugs [87] Karumanchi SA, Jiang L, Knebelmann B, Stuart-Tilley AK,
2017;26(3):265—8. Alper SL, Sukhatme VP. VHL tumor suppressor regulates
[70] Walenta S, Mueller-Klieser WF. Lactate: mirror and motor Cl− /HCO3 − exchange and Na+ /H+ exchange activities in renal
of tumor malignancy. Semin Radiat Oncol 2004;14(3): carcinoma cells. Physiol Genomics 2001;5(3):119—28.
267—74. [88] Walker NM, Simpson JE, Yen PF, Gill RK, Rigsby EV, Brazill JM,
[71] Walenta S, Voelxen NF, Mueller-Klieser W. Lactate—–an inte- et al. Down-regulated in adenoma Cl/HCO3 exchanger couples
grative mirror of cancer metabolism. Recent Results Cancer with Na/H exchanger 3 for NaCl absorption in murine small
Res 2016;207:23—37. intestine. Gastroenterology 2008;135(5):1645—53, e3.
[72] Brizel DM, Schroeder T, Scher RL, Walenta S, Clough RW, [89] Sautin YY, Lu M, Gaugler A, Zhang L, Gluck SL. Phosphatidyli-
Dewhirst MW, et al. Elevated tumor lactate concentrations nositol 3-kinase-mediated effects of glucose on vacuolar
predict for an increased risk of metastases in head-and-neck H+ -ATPase assembly, translocation, and acidification of intra-
cancer. Int J Radiat Oncol Biol Phys 2001;51(2):349—53. cellular compartments in renal epithelial cells. Mol Cell Biol
[73] Walenta S, Wetterling M, Lehrke M, Schwickert G, Sundfør 2005;25(2):575—89.
K, Rofstad EK, et al. High lactate levels predict likelihood of [90] Reshkin SJ, Cardone RA, Harguindey S. Na+ -H+ exchanger, pH
metastases, tumor recurrence, and restricted patient survival regulation and cancer. Recent Patents Anticancer Drug Discov
in human cervical cancers. Cancer Res 2000;60(4):916—21. 2013;8(1):85—99.
[74] Martinez-Outschoorn UE, Lisanti MP, Sotgia F. Catabolic [91] Stock C, Mueller M, Kraehling H, Mally S, Noël J, Eder C, et al.
cancer-associated fibroblasts transfer energy and biomass to pH nanoenvironment at the surface of single melanoma cells.
anabolic cancer cells, fueling tumor growth. Semin Cancer Cell Physiol Biochem 2007;20(5):679—86.
Biol 2014;25:47—60. [92] Krähling H, Mally S, Eble JA, Noël J, Schwab A, Stock C.
[75] Whitaker-Menezes D, Martinez-Outschoorn UE, Lin Z, Ertel A, The glycocalyx maintains a cell surface pH nanoenvironment
Flomenberg N, Witkiewicz AK, et al. Evidence for a stromal- crucial for integrin-mediated migration of human melanoma
epithelial lactate shuttle in human tumors: MCT4 is a marker cells. Pflugers Arch Eur J Physiol 2009;458(6):1069—83.
of oxidative stress in cancer-associated fibroblasts. Cell Cycle [93] Opavský R, Pastoreková S, Zelnik V, Gibadulinova A, Stan-
2011;10(11):1772—83. bridge EJ, Závada J, et al. Human MN/CA9 gene, a novel
[76] Sanità P, Capulli M, Teti A, Galatioto GP, Vicentini C, Chiarugi member of the carbonic anhydrase family: structure and exon
P, et al. Tumor-stroma metabolic relationship based on lactate to protein domain relationships. Genomics 1996;33(3):480—7.
shuttle can sustain prostate cancer progression. BMC Cancer [94] Mahon BP, Pinard MA, McKenna R. Targeting car-
2014;14:154. bonic anhydrase IX activity and expression. Molecules
[77] Offermanns S. Free fatty acid (FFA) and hydroxy car- 2015;20(2):2323—48.
boxylic acid (HCA) receptors. Annu Rev Pharmacol Toxicol [95] Kaluz S, Kaluzová M, Liao SY, Lerman M, Stanbridge EJ. Tran-
2014;54:407—34. scriptional control of the tumor- and hypoxia-marker carbonic
[78] Lee YJ, Shin KJ, Park SA, Park KS, Park S, Heo K, et al. G- anhydrase 9: A one transcription factor (HIF-1) show? Biochim
protein-coupled receptor 81 promotes a malignant phenotype Biophys Acta 2009;1795(2):162—72.
Cancer metabolism 19
[96] Barathova M, Takacova M, Holotnakova T, Gibadulinova A, cer cells use oxidative mitochondrial metabolism. Cell Cycle
Ohradanova A, Zatovicova M, et al. Alternative splicing vari- 2010;9(17):3506—14.
ant of the hypoxia marker carbonic anhydrase IX expressed [113] Salem AF, Howell A, Sartini M, Sotgia F, Lisanti MP. Downreg-
independently of hypoxia and tumour phenotype. Br J Cancer ulation of stromal BRCA1 drives breast cancer tumor growth
2008;98(1):129—36. via upregulation of HIF-1␣, autophagy and ketone body pro-
[97] Chiche J, Ilc K, Laferrière J, Trottier E, Dayan F, Mazure duction. Cell Cycle 2012;11(22):4167—73.
NM, et al. Hypoxia-inducible carbonic anhydrase IX and [114] Stilling RM, van de Wouw M, Clarke G, Stanton C, Dinan
XII promote tumor cell growth by counteracting acidosis TG, Cryan JF. The neuropharmacology of butyrate: the bread
through the regulation of the intracellular pH. Cancer Res and butter of the microbiota-gut-brain axis. Neurochem Int
2009;69(1):358—68. 2016;99:110—32.
[98] Choschzick M, Woelber L, Hess S, zu Eulenburg C, Schwarz [115] Scheck AC, Abdelwahab MG, Fenton KE, Stafford P. The keto-
J, Simon R, et al. Overexpression of carbonic anhydrase IX genic diet for the treatment of glioma: insights from genetic
(CAIX) in vulvar cancer is associated with tumor progression profiling. Epilepsy Res 2012;100(3):327—37.
and development of locoregional lymph node metastases. Vir- [116] Schwartz K, Chang HT, Nikolai M, Pernicone J, Rhee S,
chows Arch 2010;456(5):483—90. Olson K, et al. Treatment of glioma patients with ketogenic
[99] Ilie M, Mazure NM, Hofman V, Ammadi RE, Ortholan C, Bon- diets: report of two cases treated with an IRB-approved
netaud C, et al. High levels of carbonic anhydrase IX in energy-restricted ketogenic diet protocol and review of the
tumour tissue and plasma are biomarkers of poor prognos- literature. Cancer Metab 2015;3(3).
tic in patients with non-small cell lung cancer. Br J Cancer [117] Woolf EC, Scheck AC. The ketogenic diet for the treatment of
2010;102(11):1627—35. malignant glioma. J Lipid Res 2015;56(1):5—10.
[100] Chen Y, Li X, Wu S, Xu G, Zhou Y, Gong L, et al. Expression of [118] Abdelwahab MG, Fenton KE, Preul MC, Rho JM, Lynch A,
HIF-1␣ and CAIX in nasopharyngeal carcinoma and their corre- Stafford P, et al. The ketogenic diet is an effective adjuvant
lation with patients’ prognosis. Med Oncol 2014;31(12):304. to radiation therapy for the treatment of malignant glioma.
[101] Zatovicova M, Sedlakova O, Svastova E, Ohradanova A, PLoS One 2012;7(5):e36197.
Ciampor F, Arribas J, et al. Ectodomain shedding of the [119] Shimazu T, Hirschey MD, Newman J, He W, Shirakawa
hypoxia-induced carbonic anhydrase IX is a metalloprotease- K, Le Moan N, et al. Suppression of oxidative stress
dependent process regulated by TACE/ADAM17. Br J Cancer by -hydroxybutyrate, an endogenous histone deacetylase
2005;93(11):1267—76. inhibitor. Science 2013;339(6116):211—4.
[102] Rosenberg V, Pastorekova S, Zatovicova M, Vidlickova I, Jelen- [120] Woolf EC, Curley KL, Liu Q, Turner GH, Charlton JA, Preul
ska L, Slezak P. High serum carbonic anhydrase IX predicts MC, et al. The ketogenic diet alters the hypoxic response
shorter survival in head and neck cancer. Bratisl Lek Listy and affects expression of proteins associated with angiogen-
2016;117(4):201—4. esis, invasive potential and vascular permeability in a mouse
[103] Smith AD, Truong M, Bristow R, Yip P, Milosevic MF, Joshua glioma model. PLoS One 2015;10(6):e0130357.
AM. The utility of serum CA9 for prognostication in prostate [121] Bourguignon LY, Singleton PA, Diedrich F, Stern R, Gilad E.
cancer. Anticancer Res 2016;36(9):4489—92. CD44 interaction with Na+ -H+ exchanger (NHE1) creates acidic
[104] Carney WP. Circulating oncoproteins HER2/neu, EGFR and microenvironments leading to hyaluronidase-2 and cathep-
CAIX (MN) as novel cancer biomarkers. Expert Rev Mol Diagn sin B activation and breast tumor cell invasion. J Biol Chem
2007;7(3):309—19. 2004;279(26):26991—7007.
[105] Papworth K, Sandlund J, Grankvist K, Ljungberg B, Rasmuson [122] Poola I, Abraham J, Marshalleck JJ, Yue Q, Lokeshwar VB,
T. Soluble carbonic anhydrase IX is not an independent prog- Bonney G, et al. Molecular risk assessment for breast cancer
nostic factor in human renal cell carcinoma. Anticancer Res development in patients with ductal hyperplasias. Clin Cancer
2010;30(7):2953—7. Res 2008;14(4):1274—80.
[106] Gigante M, Li G, Ferlay C, Perol D, Blanc E, Paul S, et al. Prog- [123] Madan AK, Yu K, Dhurandhar N, Cullinane C, Pang Y, Beech
nostic value of serum CA9 in patients with metastatic clear DJ. Association of hyaluronidase and breast adenocarcinoma
cell renal cell carcinoma under targeted therapy. Anticancer invasiveness. Oncol Rep 1999;6(3):607—9.
Res 2012;32(12):5447—51. [124] Sun X, Zhao D, Li YL, Sun Y, Lei XH, Zhang JN, et al.
[107] Woelber L, Mueller V, Eulenburg C, Schwarz J, Carney W, Regulation of ASIC1 by Ca2+ /calmodulin-dependent protein
Jaenicke F, et al. Serum carbonic anhydrase IX during first-line kinase II in human glioblastoma multiforme. Oncol Rep
therapy of ovarian cancer. Gynecol Oncol 2010;117(2):183—8. 2013;30(6):2852—8.
[108] Fidan E, Mentese A, Ozdemir F, Deger O, Kavgaci H, Caner [125] Zhang Y, Zhang T, Wu C, Xia Q, Xu D. ASIC1a mediates
Karahan S, et al. Diagnostic and prognostic significance of CA the drug resistance of human hepatocellular carcinoma
IX and suPAR in gastric cancer. Med Oncol 2013;30(2):540. via the Ca2+ /PI3-kinase/AKT signaling pathway. Lab Invest
[109] Hektoen HH, Flatmark K, Andersson Y, Dueland S, Redalen KR, 2017;97(1):53—69.
Ree AH. Early increase in circulating carbonic anhydrase IX [126] Laplante M. Sabatini DM: mTOR signaling in growth control
during neoadjuvant treatment predicts favourable outcome and disease. Cell 2012;149(2):274—93.
in locally advanced rectal cancer. BMC Cancer 2015;15:543. [127] Kato Y, Nakayama Y, Umeda M, Miyazaki K. Induction of
[110] Carvalho S, Troost EG, Bons J, Menheere P, Lambin P, Oberije 103-kDa gelatinase/type IV collagenase by acidic culture con-
C. Prognostic value of blood-biomarkers related to hypoxia, ditions in mouse metastatic melanoma cell lines. J Biol Chem
inflammation, immune response and tumour load in non-small 1992;267(16):11424—30.
cell lung cancer—–a survival model with external validation. [128] Kato Y, Ozono S, Shuin T, Miyazaki K. Slow induction of gelati-
Radiother Oncol 2016;119(3):487—94. nase B mRNA by acidic culture conditions in mouse metastatic
[111] Dunger DB, Sperling MA, Acerini CL, Bohn DJ, Daneman D, melanoma cells. Cell Biol Int 1996;20(5):375—7.
Danne TP, et al. ESPE/LWPES consensus statement on dia- [129] Kato Y, Lambert CA, Colige AC, Mineur P, Noël A,
betic ketoacidosis in children and adolescents. Arch Dis Child Frankenne F, et al. Acidic extracellular pH induces
2004;89(2):188—94. matrix metalloproteinase-9 expression in mouse
[112] Bonuccelli G, Tsirigos A, Whitaker-Menezes D, Pavlides S, metastatic melanoma cells through the phospholipase
Pestell RG, Chiavarina B, et al. Ketones and lactate fuel
tumor growth and metastasis: evidence that epithelial can-
20 Y. Kato et al.
D-mitogen-activated protein kinase signaling. J Biol Chem [147] Ganea E, Harding JJ. Glutathione-related enzymes and the
2005;280(12):10938—44. eye. Curr Eye Res 2006;31(1):1—11.
[130] Peppicelli S, Bianchini F, Torre E, Calorini L. Contribution of [148] Ying W. NAD+ /NADH and NADP+ /NADPH in cellular func-
acidic melanoma cells undergoing epithelial-to-mesenchymal tions and cell death: regulation and biological consequences.
transition to aggressiveness of non-acidic melanoma cells. Antioxid Redox Signal 2008;10(2):179—206.
Clin Exp Metastasis 2014;31(4):423—33. [149] Son J, Lyssiotis CA, Ying H, Wang X, Hua S, Ligorio M, et al.
[131] Riemann A, Schneider B, Ihling A, Nowak M, Sauvant C, Thews Glutamine supports pancreatic cancer growth through a KRAS-
O, et al. Acidic environment leads to ROS-induced MAPK sig- regulated metabolic pathway. Nature 2013;496(7443):101—5.
naling in cancer cells. PLoS One 2011;6(7):e22445. [150] Alfarouk KO. Tumor metabolism, cancer cell transporters, and
[132] Kato Y, Ozawa S, Tsukuda M, Kubota E, Miyazaki K, St-Pierre microenvironmental resistance. J Enzyme Inhib Med Chem
Y, et al. Acidic extracellular pH increases calcium influx- 2016;31(6):859—66.
triggered phospholipase D activity along with acidic sphin- [151] Ushio-Fukai M, Nakamura Y. Reactive oxygen species and
gomyelinase activation to induce matrix metalloproteinase- angiogenesis: NADPH oxidase as target for cancer therapy.
9 expression in mouse metastatic melanoma. FEBS J Cancer Lett 2008;266(1):37—52.
2007;274(12):3171—83. [152] Tew KD. Glutathione-associated enzymes in anticancer drug
[133] Maeda T, Yuzawa S, Suzuki A, Baba Y, Nishimura Y, Kato Y. RhoA resistance. Cancer Res 1994;54(16):4313—20.
mediates the expression of acidic extracellular pH-induced [153] Riemann A, Schneider B, Gündel D, Stock C, Gekle M, Thews O.
matrix metalloproteinase-9 mRNA through phospholipase D1 Acidosis Promotes Metastasis Formation by Enhancing Tumor
in mouse metastatic B16-BL6 melanoma cells. Int J Oncol Cell Motility. Adv Exp Med Biol 2016;876:215—20.
2016;48(3):1251—7. [154] Riemann A, Ihling A, Schneider B, Gekle M, Thews O. Impact
[134] Fang Y, Vilella-Bach M, Bachmann R, Flanigan A, Chen J. of extracellular acidosis on intracellular pH control and cell
Phosphatidic acid-mediated mitogenic activation of mTOR sig- signaling in tumor cells. Adv Exp Med Biol 2013;789:221—8.
naling. Science 2001;294(5548):1942—5. [155] Hiramoto K, Satoh H, Suzuki T, Moriguchi T, Pi J, Shimosegawa
[135] Hui L, Abbas T, Pielak RM, Joseph T, Bargonetti J, Foster T, et al. Myeloid lineage-specific deletion of antioxidant sys-
DA. Phospholipase D elevates the level of MDM2 and sup- tem enhances tumor metastasis. Cancer Prev Res (Phila)
presses DNA damage-induced increases in p53. Mol Cell Biol 2014;7(8):835—44.
2004;24(13):5677—86. [156] Le Gal K, Ibrahim MX, Wiel C, Sayin VI, Akula MK, Karlsson
[136] Suzuki A, Maeda T, Baba Y, Shimamura K, Kato Y. Acidic extra- C, et al. Antioxidants can increase melanoma metastasis in
cellular pH promotes epithelial mesenchymal transition in mice. Sci Transl Med 2015;7(308):308re8.
Lewis lung carcinoma model. Cancer Cell Int 2014;14(1):129. [157] Piskounova E, Agathocleous M, Murphy MM, Hu Z, Huddlestun
[137] Peppicelli S, Bianchini F, Toti A, Laurenzana A, Fibbi G, SE, Zhao Z, et al. Oxidative stress inhibits distant metastasis
Calorini L. Extracellular acidity strengthens mesenchymal by human melanoma cells. Nature 2015;527(7577):186—91.
stem cells to promote melanoma progression. Cell Cycle [158] Wang H, Liu X, Long M, Huang Y, Zhang L, Zhang R, et al.
2015;14(19):3088—100. NRF2 activation by antioxidant antidiabetic agents acceler-
[138] Dienstmann R, Rodon J, Serra V, Tabernero J. Picking the ates tumor metastasis. Sci Transl Med 2016;8(334):334ra51.
point of inhibition: a comparative review of PI3K/AKT/mTOR [159] Cairns RA, Harris IS, Mak TW. Regulation of cancer cell
pathway inhibitors. Mol Cancer Ther 2014;13(5):1021—31. metabolism. Nat Rev Cancer 2011;11(2):85—95.
[139] Vinnars E, Bergstöm J, Fürst P. Influence of the postoperative [160] Jewell JL, Russell RC, Guan KL. Amino acid signalling
state on the intracellular free amino acids in human muscle upstream of mTOR. Nat Rev Mol Cell Biol 2013;14(3):133—9.
tissue. Ann Surg 1975;182(6):665—71. [161] Chong ZZ, Maiese K. Mammalian target of rapamycin signal-
[140] Altman BJ, Stine ZE, Dang CV. From Krebs to clinic: glu- ing in diabetic cardiovascular disease. Cardiovasc Diabetol
tamine metabolism to cancer therapy. Nat Rev Cancer 2012;11:45.
2016;16(10):619—34. [162] Shimobayashi M, Hall MN. Multiple amino acid sensing inputs
[141] Matés JM, Segura JA, Martín-Rufián M, Campos-Sandoval JA, to mTORC1. Cell Res 2016;26(1):7—20.
Alonso FJ, Márquez J. Glutaminase isoenzymes as key regu- [163] Dekanty A, Lavista-Llanos S, Irisarri M, Oldham S, Wappner P.
lators in metabolic and oxidative stress against cancer. Curr The insulin-PI3K/TOR pathway induces a HIF-dependent tran-
Mol Med 2013;13(4):514—34. scriptional response in Drosophila by promoting nuclear local-
[142] Hartman SC. Purines and pyrimidines. In: Greenberg DM, edi- ization of HIF-␣/Sima. J Cell Sci 2005;118(Pt. 23):5431—41.
tor. Metaboric pathways. New York and London: Academic [164] Nemazanyy I, Espeillac C, Pende M, Panasyuk G. Role of
Press; 1970. p. 1—68. PI3K, mTOR and Akt2 signalling in hepatic tumorigenesis
[143] Cetinbas NM, Sudderth J, Harris RC, Cebeci A, Negri GL, Yil- via the control of PKM2 expression. Biochem Soc Trans
maz OH, et al. Glucose-dependent anaplerosis in cancer cells 2013;41(4):917—22.
is required for cellular redox balance in the absence of glu- [165] Cheng SC, Quintin J, Cramer RA, Shepardson KM, Saeed S,
tamine. Sci Rep 2016;6:32606. Kumar V, et al. mTOR- and HIF-1␣-mediated aerobic gly-
[144] Wise DR, DeBerardinis RJ, Mancuso A, Sayed N, Zhang colysis as metabolic basis for trained immunity. Science
XY, Pfeiffer HK, et al. Myc regulates a transcriptional 2014;345(6204):1250684.
program that stimulates mitochondrial glutaminolysis and [166] Miyazaki M, Miyazaki K, Chen S, Chandra V, Wagatsuma
leads to glutamine addiction. Proc Natl Acad Sci U S A K, Agata Y, et al. The E-Id protein axis modulates the
2008;105(48):18782—7. activities of the PI3K-AKT-mTORC1-Hif1a and c-myc/p19Arf
[145] Townsend DM, Tew KD. The role of glutathione-S- pathways to suppress innate variant TFH cell development,
transferase in anti-cancer drug resistance. Oncogene thymocyte expansion, and lymphomagenesis. Genes Dev
2003;22(47):7369—75. 2015;29(4):409—25.
[146] Neumann C, Boubakari Grünert R, Bednarski PJ. Nicotinamide [167] Xu BH, Li XX, Yang Y, Zhang MY, Rao HL, Wang HY, et al. Aber-
adenine dinucleotide phosphate-regenerating system coupled rant amino acid signaling promotes growth and metastasis of
to a glutathione-reductase microtiter method for determina- hepatocellular carcinomas through Rab1A-dependent activa-
tion of total glutathione concentrations in adherent growing tion of mTORC1 by Rab1A. Oncotarget 2015;6(25):20813—28.
cancer cell lines. Anal Biochem 2003;320(2):170—8.
Cancer metabolism 21
[168] Jung CH, Ro SH, Cao J, Otto NM. Kim DH mTOR regulation of [179] Kurth I, Hein L, Mäbert K, Peitzsch C, Koi L, Cojoc M,
autophagy. FEBS Lett 2010;584(7):1287—95. et al. Cancer stem cell related markers of radioresistance
[169] Sancak Y, Peterson TR, Shaul YD, Lindquist RA, Thoreen in head and neck squamous cell carcinoma. Oncotarget
CC, Bar-Peled L, et al. The Rag GTPases bind raptor 2015;6(33):34494—509.
and mediate amino acid signaling to mTORC1. Science [180] Habu N, Imanishi Y, Kameyama K, Shimoda M, Tokumaru Y,
2008;320(5882):1496—501. Sakamoto K, et al. Expression of Oct3/4 and Nanog in the
[170] Budanov AV. Karin M: p53 target genes sestrin1 and ses- head and neck squamous carcinoma cells and its clinical impli-
trin2 connect genotoxic stress and mTOR signaling. Cell cations for delayed neck metastasis in stage I/II oral tongue
2008;134(3):451—60. squamous cell carcinoma. BMC Cancer 2015;15:730.
[171] Zhou G, Wang J, Zhao M, Xie TX, Tanaka N, Sano D, et al. [181] Prochazka L, Tesarik R, Turanek J. Regulation of alternative
Gain-of-function mutant p53 promotes cell growth and cancer splicing of CD44 in cancer. Cell Signal 2014;26(10):2234—9.
cell metabolism via inhibition of AMPK activation. Mol Cell [182] Takayama T, Kubo T, Morikawa A, Morita T, Nagano O, Saya
2014;54(6):960—74. H. Potential of sulfasalazine as a therapeutic sensitizer for
[172] Ganley IG, Lam du H, Wang J, Ding X, Chen S, Jiang X. ULK1. CD44 splice variant 9-positive urogenital cancer. Med Oncol
ATG13. FIP200 complex mediates mTOR signaling and is essen- 2016;33(5):45.
tial for autophagy. J Biol Chem 2009;284(18):12297—305. [183] Seishima R, Okabayashi K, Nagano O, Hasegawa H, Tsuruta
[173] Egan D, Kim J, Shaw RJ, Guan KL. The autophagy initiat- M, Shimoda M, et al. Sulfasalazine, a therapeutic agent for
ing kinase ULK1 is regulated via opposing phosphorylation by ulcerative colitis, inhibits the growth of CD44v9+ cancer stem
AMPK and mTOR. Autophagy 2011;7(6):643—4. cells in ulcerative colitis-related cancer. Clin Res Hepatol Gas-
[174] Venneti S, Dunphy MP, Zhang H, Pitter KL, Zanzonico P, troenterol 2016;40(4):487—93.
Campos C, et al. Glutamine-based PET imaging facilitates [184] Wada T, Ishimoto T, Seishima R, Tsuchihashi K, Yoshikawa M,
enhanced metabolic evaluation of gliomas in vivo. Sci Transl Oshima H, et al. Functional role of CD44v-xCT system in the
Med 2015;7(274):274ra17. development of spasmolytic polypeptide-expressing metapla-
[175] Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres- sia. Cancer Sci 2013;104(10):1323—9.
Cortes J, et al. A cell initiating human acute myeloid [185] Shitara K, Doi T, Nagano O, Imamura CK, Ozeki T, Ishii Y, et al.
leukaemia after transplantation into SCID mice. Nature Dose-escalation study for the targeting of CD44v+ cancer stem
1994;367(6464):645—8. cells by sulfasalazine in patients with advanced gastric cancer
[176] Bonnet D, Dick JE. Human acute myeloid leukemia is (EPOC 1205). Gastric Cancer 2017;20(2):341—9.
organized as a hierarchy that originates from a primitive [186] Iliopoulos D, Hirsch HA, Wang G, Struhl K. Inducible formation
hematopoietic cell. Nat Med 1997;3(7):730—7. of breast cancer stem cells and their dynamic equilibrium with
[177] Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke non-stem cancer cells via IL6 secretion. Proc Natl Acad Sci U
MF. Prospective identification of tumorigenic breast cancer S A 2011;108(4):1397—402.
cells. Proc Natl Acad Sci U S A 2003;100(7):3983—8.
[178] Jamieson CH, Ailles LE, Dylla SJ, Muijtjens M, Jones C,
Zehnder JL, et al. Granulocyte-macrophage progenitors as
candidate leukemic stem cells in blast-crisis CML. N Engl J
Med 2004;351(7):657—67.