Clinical impact of the Warburg effect in gastrointestinal cancer (Review)

  • Authors:
    • Hiroshi Sawayama
    • Takatsugu Ishimoto
    • Hidetaka Sugihara
    • Nobutomo Miyanari
    • Yuji Miyamoto
    • Yoshifumi Baba
    • Naoya Yoshida
    • Hideo Baba
  • View Affiliations

  • Published online on: July 25, 2014     https://doi.org/10.3892/ijo.2014.2563
  • Pages: 1345-1354
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cancer cells exhibit altered glucose metabolism, termed the Warburg effect, which is described by the increased uptake of glucose and the conversion of glucose to lactate in cancer cells under adequate oxygen tension. Recent genetic and metabolic analyses have provided insights into the molecular mechanisms of genes that are involved in the Warburg effect and tumorigenesis. The aim of this review was to discuss significant molecular insights into clinical impacts of the Warburg effect such as oncogenic alterations and overexpression of transcriptional factors (c-Myc and hypoxia-inducible factor), metabolite transporters (glucose transporters) and glycolytic enzymes (hexokinases 2, pyruvate kinase M2, pyruvate dehydrogenase kinase, isozyme 1, lactate dehydrogenase A). Overexpression of transcriptional factors, metabolite transporters and glycolytic enzymes was associated with poor prognosis and may be associated with chemoradiotherapy resistance in multiple gastrointestinal cancer cell types. Novel small molecules targeting these enzymes or transporters exert anti-proliferative effects. Glycolytic enzymes and metabolite transporters may be significant biomarkers for predicting cancer prognosis and may be therapeutic targets in gastrointestinal cancer.

1. Introduction

Cancers exhibit altered glucose metabolism, defined as the Warburg effect (1), which is characterized by an increased uptake of glucose (2) and the conversion of glucose to lactate in cancer cells, rather than catabolizing glucose via the TCA cycle under adequate oxygen tension (3). While the electron transfer system generates 36 ATP molecules per glucose molecule across the mitochondrial inner membrane, glycolysis metabolizes glucose to pyruvate in the cytoplasm to produce a net 2 ATP molecules from each glucose. The Warburg effect represents high levels of glycolysis and thus enables the clinical application of metabolic imaging, such as 18F-fluorodeoxyglucose positron emission tomography (FDG-PET), which is a non-invasive imaging technique that allows quantification of tumor activity on the basis of altered tissue glucose metabolism (4). Small molecule inhibitors targeting the enzymes that function in the Warburg effect have been identified and pursued in preclinical studies.

The direct mechanistic link between an activated oncogene and altered glucose metabolism is regulated by phosphoinositide 3-kinase (PI3K) (5), Akt (6), p53 (7,8), AMP-activated protein kinase (AMPK) (9,10), c-Myc and hypoxia-inducible factor (HIF). c-Myc and HIF1A transcription factors target many of the same glycolytic enzyme genes, including hexokinase 2 (HK2), pyruvate kinase type M2 (PKM2), lactate dehydrogenase A (LDHA), and pyruvate dehydrogenase kinase, isozyme 1 (PDK1). Recent investigations using genetic and metabolic analyses have provided insights into the molecular mechanisms of these genes that contribute to the Warburg effect and tumorigenesis (Fig. 1).

In this review, significant molecular insights into clinical impacts of the Warburg effect, such as oncogenic alterations and overexpression of glycolytic enzymes and metabolite transporters, will be discussed.

2. HIF-1A and c-Myc transcription factors and the Warburg effect

HIF-1A and c-Myc cooperatively induce a transcriptional program for glycolysis. HIF plays a crucial role in cellular adaptation to hypoxia and regulates the expression of genes responsible for glucose metabolism, angiogenesis, and cell survival (11). Cellular HIF levels are regulated by both an oxygen-dependent pathway and an oxygen-independent pathway. HIF contains two key regulatory subunits, HIF1A and endothelial PAS domain protein 1 (EPAS1; HIF-2), and the genes encoding these proteins are overexpressed in human cancers (12,13). Many studies have assessed the significance of HIF-1A positive expression in the prediction of clinical outcome of gastrointestinal cancer. HIF-1A expression is associated with poor prognosis in esophageal squamous cell carcinoma (ESCC) (14,15), gastric cancer (16,17), colorectal cancer (CRC) (18) and hepatocellular carcinoma (HCC) (19). Low expression of HIF1A may be associated with a favorable effect of 5-FU-based adjuvant chemotherapy in gastric cancer patients (20,21). HIF-2A is associated with poor survival in gastric cancer patients (22) but not CRC patients (18,23).

The c-Myc oncogene, a member of the MYC family, encodes the transcription factor c-Myc and is upregulated in many human cancers, linking altered cellular metabolism to tumorigenesis (24). MYC gene expressions are often elevated or deregulated in human neoplasms, and c-Myc seems to be at the crossroads of several important pathways and processes involved in carcinogenesis. MYC deregulation due to gene amplification (25), chromosomal translocation or insertion (26), mutations (27), and epigenetic modifications (28) has been reported in different types of cancers. The number of studies of MYC expression as detected by immunohistochemistry (IHC) is less than that of HIF1A. c-Myc overexpression and promoter hypomethylation may have a role in the gastric carcinogenesis process and c-Myc deregulation was associated mainly with poor prognosis (29). c-Myc expression detected by IHC was associated with poor prognosis in pancreatic cancer (30), but its expression was not associated with poor prognosis in CRC patients (18,23) (Table I).

Table I

Impact of HIF and MYC on cancer prognosis and correlation with clinicopathological features.

Table I

Impact of HIF and MYC on cancer prognosis and correlation with clinicopathological features.

TotalPrognosis


OrganN%Cut-offsExpression correlated with: (condition)UnivariateMultivariate(Ref.)
HIF-1A
 ESCC1,261Depth of invasion, N+, stage, VEGF (meta-analysis-2011)NANA(16)
 ESCC21568Scores 3–4VEGFDFS: poor
OS: NS
DFS: NS
OS: NS
(14)
 ESCC9668Score 4–6N+ (T1b patients)DSS: poor
DFS: poor
Poor(15)
 GCNA (meta-analysis 2003–2012)DFS: NS
OS: poor
NA(16)
 GC1103Differentiation, T-stage
N+, ly+, v+, stage (meta-analysis 2003–2013)
OS: poorNA(17)
 GC21639>10% HIF1A+-p53+ cases undifferentiated, ly+, N+OS: poorOS: poor(84)
 GC19352N+DFS: NS
OS: NS
DFS: poor
OS: poor
(45)
 GC12866>5%Histology, depth of invasion
VEGF expression, MVD
DFS: poor
OS: poor
DFS: poor
OS: poor
(85)
 GC6458No correlation [adjuvant CT S-1 (77%)]DFS: poor
DSS: poor
DFS: poor
DSS: NS
(21)
 GC4457>10%No correlation (adjuvant CT 5-FU based)DSS: poor
DFS: poor
NA(20)
 CRC73119>50%COX-2, CIMP-high
LINE1 hypomethylation
CSS: poor
OS: poor
CSS: poor
OS: poor
(18)
 RC9054N+, v+, stageDFS: poor
CSS: poor
OS: poor(23)
 RC9255Scaling systempT4, N+, v+ (T3,4/N+/−)DFS: poor
OS: poor
DFS: poor
OS: poor
(86)
 HCC953Tumor grade, N+, v+ (meta-analysis-2013)DFS: poor
OS: poor
(19)
 HCC110Male, LC, COX-2, PDGFRA
MMP7, MMP9, MYC
DFS: poor
OS: poor
DFS: poor
OS: poor
(87)
 HCC20063Intrahepatic metastasisDFS: poor
OS: poor
DFS: poor
OS: poor
(88)
 PC5066>5%VEGFDFS: NS
OS: NS
NA(89)
HIF-2A
 GC8038>Score 0Diffuse typeDFS: poor
OS: poor
CSS: NS
OS: NS
(22)
 CRC73119>50%Low tumor grade, male, BMI<30CSS: NS
OS: NS
CSS: NS
OS: NS
(18)
 RC9064No correlationDFS: NS
CSS: NS
(23)
MYC
 GC12577>10%Intestinal-type, late-onset deeper tumor extension, M+NANA(29)
 PC7052Score 5–9Perineural invasion, stageOS: poorOS: poor(30)
 CRC73119>50%Low tumor grade male, BMI<30CSS: NS
OS: NS
CSS: NS
OS: NS
(18)
 RC9064No correlationDFS: NS
CSS: NS
(23)

[i] ESCC, esophageal squamous cell carcinoma; GC, gastric cancer; CRC, colorectal cancer; RC, rectal cancer; HCC, hepatocellular carcinoma; PC, pancreatic cancer; NA, no assessment; NS, not significant.

3. Glucose transporters (Gluts)

Glut1 is composed of 492 amino acid residues and possesses a single site of N-linked glycosylation at N45 (31). Multiple glucose transporter-like proteins have been identified and characterized (32) with sequence similarity to Glut1, and these genes appear to belong to the family of solute carriers 2A (SLC2A, protein symbol Glut). The 14 Gluts are categorized into three classes based on sequence similarity: Class 1 (Gluts 1–4 and 14), Class 2 (Gluts 5, 7, 9 and 11), and Class 3 (Gluts 6, 8, 10, 12, and HMIT) (32). Glut families were evaluated with the GEO data set in silico (http://www.ncbi.nlm.nih.gov/gds/). Glut1 mRNA levels were remarkably upregulated in tumor lesions compared with normal lesions in CRC (GDS 4382), ESCC (GDS 3838) and pancreatic cancer (GDS 4336) (Table II). Several studies have been published on Glut family members, especially Glut3 (3335), but Glut1 has been the main focus of investigation. A previous study evaluating Glut1 by IHC in tissue microarray slides comprising 1,955 samples detected Glut1 positivity in 47% prostate adenocarcinomas, 29% thyroid cancer, 10% gastric cancer, 5% breast adenocarcinomas, 36% head and neck SCC, 42% uterine cervix SCC, 18.6% glioblastomas and 9.4% retinoblastomas (36).

Table II

Overexpression of metabolite transporters and glycolytic enzymes in the Warburg effect.

Table II

Overexpression of metabolite transporters and glycolytic enzymes in the Warburg effect.

Colorectal cancer (N=17)
GDS4382
ESCC (N=17)
GDS3838
Panctreatic cancer (N=45)
GDS4336



T/N ratio95% CIP-valueT/N ratio95% CIP-valueT/N ratio95% CIP-value
Glut11.90(1.16–3.09)0.012.44(1.78–3.34)<0.0013.58(2.74–4.67)<0.001
Glut20.92(0.87–0.98)0.010.97(0.93–1.01)NS0.60(0.46–0.80)<0.001
Glut31.55(0.72–3.31)NS1.96(1.23–3.13)0.011.54(1.18–2.01)<0.001
Glut41.18(0.99–1.40)NS0.97(0.83–1.13)NS0.89(0.83–0.97)0.01
Glut50.52(0.37–0.72)<0.0011.08(0.80–1.44)NS1.10(0.91–1.31)NS
Glut60.84(0.71–1.00)0.051.31(1.12–1.54)<0.0010.95(0.88–1.02)NS
Glut80.59(0.10–3.60)NS1.17(1.00–1.36)0.040.93(0.88–0.98)0.01
Glut91.13(1.01–1.26)0.031.21(1.01–1.45)0.041.07(0.98–1.17)NS
Glut100.65(0.35–1.19)NS0.89(0.66–1.18)NS1.13(1.02–1.25)0.02
Glut111.33(0.96–1.85)NS0.96(0.88–1.05)NS0.85(0.78–0.93)<0.001
Glut141.72(1.08–2.72)0.031.45(1.04–2.02)0.031.10(0.98–1.23)NS
HK20.46(0.26–0.79)0.0091.53(1.16–2.03)0.0052.55(1.97–3.30)<0.001
LDHA1.05(0.92–1.19)NS0.92(0.78–1.10)NS1.89(1.57–2.28)<0.001
PMK20.80(0.65–0.98)0.0331.41(1.02–1.95)0.042.03(1.72–2.40)<0.001
PDK11.18(0.85–1.63)NS1.37(1.08–1.74)0.0121.38(1.18–1.61)<0.001

Glut1 is transcriptionally regulated by c-Myc (24) and HIF1A (37). A recent study demonstrated that Glut1 was one of three genes consistently upregulated in cells with KRAS or BRAF mutations (38). Glut1 expression in CRC cells was positively correlated with FDG accumulation and KRAS/BRAF mutation (39). EGFR and ERK1/2 correlate with levels of PKM2 Ser 37 phosphorylation, and nuclear PKM2 induces c-Myc expression, resulting in the upregulation of Glut1 (40). In a recent study using xenografts, overexpression of Glut1 in a mammary tumor cell lines with low levels of endogenous Glut1 results from both a decrease in apoptosis and an increase in proliferation (41).

Glut1 expression is generally absent in normal tissue, but in multiple gastrointestinal cancer cell types, Glut1 expression is remarkably enhanced. Glut1 positivity is associated with poor prognosis in diverse gastrointestinal cancers, ESCC (15,42,43), gastric cancer (44,45), CRC (46,47), pancreatic cancer, HCC (48), and gallbladder cancer (49,50) (Table III).

Table III

Impact of Glut1 and glycolytic enzymes on prognosis and correlation with clinicopathological features.

Table III

Impact of Glut1 and glycolytic enzymes on prognosis and correlation with clinicopathological features.

TotalPrognosis


OrganN%Cut-offsExpression correlated with: (condition)UnivariateMultivariate(Ref.)
Glut1
 ESCC14543>50%pT3, v+ MVD (no preoperative treatment)DFS: poor
CSS: poor
DFS: NS
CCS: NS
(42)
 ESCC6348>30%No correlation (curative operation)OS: poorOS: NS(43)
 ESCC9671Score 4–6N+ (T1b patients)DFS: poor
CSS: poor
NS(15)
 GC61730>1%pap>por or tub, T-stage
N+, ly+, v+, H+, stage
OS: poorOS: poor(44)
 GC15224>30%T2-T4, N+, diffuse typeDFS: NS
OS: NS
DFS: NS
OS: NS
(70)
 GC19343Age >65, T2-T4, N+, stage, intestinal typeOS: poorOS: NS(45)
 CRC163Poorly differentied higher in stage III + IVOS: poorOS: poor(90)
 CRC11218>50%N+,CSS: poorCSS: poor(46)
 RC4648>10%No correlationDFS: p=0.066NA(47)
 PC9475>50%Historogical grade, MIB1 (ductal AC)OS: poorOS: poor(91)
 HCC6337Scoring
≥Score 1
SUV, TNR, Ki67LIDFS: poor
OS: poor
NA(48)
 GB5634>50%Perinecrotic areasOS: poorNA(49)
 GB7152Histologic tumor type tumor stageOS: poor(50)
HK2
 GC25717>30%No correlationDFS: NS
OS: NS
DFS: NS
OS: NS
(92)
 GC1525>30%No correlationDFS: NS
OS: NS
DFS: NS
OS: NS
(70)
 GC18821Size, lower differentiation, stage, HIF1AOS: poorOS: poor(93)
 HCC15715High mod.Moderately and poorly, advanced stageOS: poorOS: poor(58)
 HCC9756No correlationOS: poorNA(94)
 HCC3181Moderately and poorly differentiatedOS: NSOS: NS(59)
PKM2
 ESCC18080IRS strong mod.Differntiation poorly tumor size, stageOS: poorOS: poor(95)
 GC36839>25%Age, t-stage, well differentiatiedOS: NSOS: NS(96)
 GC7918>25%Subgroup analysis above study (signet cell)OS: poorOS: poor(96)
 GB8056>25%Differntiation poorly, tumor size, stage, N+OS: poorOS: poor(97)
PDK-1
 GC15212>30%T3-T4, N+, tumor size
HIF-1A
DFS: poor
OS: poor
DFS: poor
OS: poor
(70)
 CC74-Blot densityExpression PDK1 deceased in cancer tissueNANA(70)
PDK-3
 CC20686 Stain+Stage, HIF-1ADFS: poor
OS: poor
NA(71)
LDH-5
 GC9462Advanced tumor, v+
HIF-1A, VEGF, COC-2
DFS: poor
OS: poor
NA(98)
 CC12877Poor differentiation
HIF1A, pKDR
DFS: poor
OS: poor
NA(78)

[i] ESCC, esophageal squamous cell carcinoma; GC, gastric cancer; CRC, colorectal cancer; RC, rectal cancer; HCC, hepatocellular carcinoma; PC, pancreatic cancer; GB, gallbladder cancer; MVD, microvessel density; ductal AC, ductal adenocarcinoma; NA, no assessment; NS, not significant.

Glut1 expression has the potential to serve as a biomarker for cancer. Anticancer therapies, such as radiation and several chemotherapeutic drugs, induce oxidative stress in targeted cells. Reactive oxygen species (ROS) are required for the fixation of radiation-induced DNA damage (51). Therefore, an accumulation of antioxidants (e.g., lactate) may induce or enhance resistance to radiation and may cause chemoresistance (52). Glut1 positivity was associated with tumor regression grade (TRG) and may be a useful predictive marker of response to chemoradiotherapy in rectal cancer (47,53).

Phloretin, a natural product found in apples and pears with Glut inhibitory activity, exerts antitumor effects in HCC and color cancer cell lines (54,55). The WZB117 small molecule inhibitor of Glut1 was effective in inhibiting cancer cell growth both in vitro and in vivo (56) (Table IV).

Table IV

Anti-proliferative effect of inhibitors of metabolite transporters and glycolytic enzymes.

Table IV

Anti-proliferative effect of inhibitors of metabolite transporters and glycolytic enzymes.

TargetInhibitorCancer type (cell lines)Dose in vitroDose in vivoCombination or drug resistance(Ref.)
Glut1WZB117LC (A549)10 μM10 mg/kg (i.p.) dailyNA(56)
PhloretinCRC (SW620)50 μMNADNR(54)
Glut2PhloretinHCC (HepG2)200 μM10 mg/kg (i.p.)
3 times per week
DNR(55)
HK-23-BrPAHCC (Huh-7)100 μM1 mg/kg (i.p.)PDI(99)
3-BrPACRC (HCT116, HT29)30 μMNAOx-resistant cells(100)
PKM-2Compound 3LC (H1299), hematopoetic (FL5.12)30 μMNAGefitinib(101)
PDKDCAHCC (Huh-7)30 mM100 mg per kg bw per daySorafinib-resistant cells(74)
DCAGC (MKN45, AGS)20 mMNA5-FU(70)
DCACRC (SW620, LoVo, LS174t, HT29)10 mMNA5-FU(73)
BX-320CRC (HCT116)
PC (MiaPaCa)
0.28 μM
0.33 μM
NANA(102)
LDHAFX11Lymphoma (P493)9 μM42 μg (i.p.) dailyFK866(81)
OxmateCRC (CT26)9 μMNAPhenformin(82)

[i] DCA, dichloroacetate; 3-BrPA, 3-bromopyruvate; DNR, daunorubicin; PDI, protein disulfide isomerase; Ox, oxaliplatin; LC, lung cancer; CRC, colorectal cancer; HCC, hepatocellular carcinoma; GC, gastric cancer; NA, no assessment; NS, not significant.

4. Glycolytic enzymes (HK2, PKM2, PDK1 and LDHA)

Hexokinases catalyze the phosphorylation of glucose to glucose-6-phosphate. This is the first and rate-limiting step in glucose metabolism. HK2 is one of four members of the hexokinase family. The four isoenzymes (HK1, HK2, HK3, and glucokinase) are structurally similar, but only HK1 and HK2 are functionally similar. HK2, but not HK1, is overexpressed in several cancer types compared with normal tissue. HK2 localizes to the outer membrane of the mitochondria and is the major hexokinase isoform expressed in cancer cells (57). High expression of HK2 confers a poor prognosis in HCC and gastric cancer (Table II), and HK2 positivity was associated with poor differentiation and advanced stage in HCC (58,59). Tumor differentiation in HCC correlated with FDG uptake (60), and the cellular retention of FDG may be mediated by HK2 (58).

The widely used 3-bromopyruvate (3-BrPA) (61) depletes cellular ATP. A previous study showed that 3-BrPA inhibits HK2 expression and exhibits anti-proliferative effects combined with daunorubicin in CRC cell lines (54) and combined with protein disulfide isomerase in HCC cell lines in vivo (55).

Pyruvate kinase (PK) is a glycolytic enzyme that catalyzes a reaction generating pyruvate and ATP from phosphoenolpyruvate (PEP) and ADP. Four isoforms of PK (L, R, M1, and M2) have been identified in mammals. Splicing of PKM is controlled by splicing repressors, and the expression of the repressors is upregulated by c-Myc oncoprotein (62,63). M2 is expressed in embryonic cells, adult stem cells, and cancer cells and is necessary for aerobic glycolysis. This metabolic phenotype provides a selective growth advantage for tumor cells in vivo (64,65). PKM2 expression is associated with poor prognosis in ESCC, gallbladder cancer and signet ring cell carcinoma of gastric cancer (Table III). Small molecule inhibitors that selectively target PKM2 have been identified, suggesting that inhibition of PKM2 could be synergistic with other targeted therapies, including gefitinib. However, small molecule activation of PKM2 that promotes PKM2 tetramer formation interferes with anabolic metabolism and suppresses tumorigenesis (66). Mutation of the ERK-phosphorylation site S37 in PKM2 blocked translocation of PKM2 to the nucleus (40), suggesting that PKM2 moves into the nucleus as a monomer. Tumor cells have multiple ways to regulate PKM2 that are favorable to cell growth and survival, including PKM2 expression, localization, post-translational modification, and allosteric regulation. PKM2 also regulates non-metabolic functions as a transcriptional coactivator and protein kinase. PKM2 is considered an attractive target for cancer treatment (67). Further studies are needed before inhibitors and activators of PKM2 can be used as therapeutic interventions (68).

PDK regulates the mitochondrial gatekeeper pyruvate dehydrogenase (PDH), which links glycolysis to the TCA cycle by reversible phosphorylation. Phosphorylation of PDH by PDK inhibits the action of PDH and halts pyruvate use in the TCA cycle (69). Four PDK isoforms have been verified in human tissue, and the expressions of the isoforms are organ specific. PDK-1 positivity was associated with poor prognosis in gastric cancer (70), but expression of PDK-1 was decreased in colon cancer compared with normal tissue. PDK-3 expression was detected in colon cancer, and PDK-3 positivity was associated with poor prognosis (71). Several studies reported the relationship between PDK positivity and prognosis in gastrointestinal cancer, but the clinical significance of PDK expression has remained unclear. Many small molecule PDK-1 inhibitors have been identified (72). DCA, a PDK-1 inhibitor, reduced lactate production and increased responsiveness to 5-FU in MKN45 cells (70) and CRC cell lines (73). DCA treatment exerts anti-proliferative effects and sorafenib resistance in HCC cell lines in vivo (74).

Lactate dehydrogenase is a tetrameric enzyme comprising two major subunits, A and/or B, resulting in five isozymes (A4, A3B1, A2B2, A1B3 and B4) that can catalyze the forward and backward conversion of pyruvate to lactate. LDHA (LDH-5, MLDH, or A4), which is the predominant form in skeletal muscle, kinetically favors the conversion of pyruvate to lactate, controlling the conversion of pyruvate to lactate in the cellular glycolytic process (75). Many human cancers have higher LDHA levels than normal tissues (76). LDHA is specifically phosphorylated at Y10 in various cancer cell lines, head and neck SCC, lung cancer, breast cancer, and prostate cancer cells and by diverse oncogenic tyrosine kinases, including FGFR1, ABL, JAK2, and FLT3 (77).

LDHA reduction can suppress the tumorigenicity of intestinal- type gastric cancer (ITGC) cells, colon cancer (78) and HCC (79). A previous study of 661 ITGC specimens showed that low LDHA expression exhibited better overall survival than high LDHA expression (80).

Similar to small interfering RNA (siRNA) reduction of LDHA expression, the FX11 small molecule inhibitor for LDHA could increase cellular oxygen consumption, increase ROS production, and induce cell death that could be partially rescued by the antioxidant NAC in a lymphoma cell line (81). Oxmate, a lactate dehydrogenase inhibitor, combined with phenformin exhibited cytotoxic effects in diverse cancer cell lines, including colon cancer (82).

5. Conclusions and future perspectives

This review describes the significance of protein expression of the transcriptional factors, glycolytic enzymes and metabolite transporters involved in the Warburg effect as potential biomarkers. The functional and therapeutic importance of the Warburg effect is increasingly recognized, and glycolysis has become a target of anticancer strategies. Novel small molecule inhibitors targeting enzymes that function in the Warburg effect have been developed and anti-proliferative effects on diverse cancer cells have been demonstrated. The gene expressions of molecular factors involved in the Warburg effect are associated with poor prognosis and may be associated with chemoradiotherapy resistance in gastrointestinal cancers. Novel small molecules exert anti-proliferative effects and may reduce chemoradiotherapy resistance in gastrointestinal cancer, breast cancer (83) and lung cancer (56) (Table IV).

Future studies should examine whether inhibitors of glycolytic enzymes and metabolite transporters are useful in gastrointestinal cancer and evaluate adverse effect and feasibility for clinical practice. Furthermore, validation of imaging techniques, which establish glucose metabolism and predict response to drugs, is required for optimal patient selection.

References

1 

Warburg O: On respiratory impairment in cancer cells. Science. 124:269–270. 1956.PubMed/NCBI

2 

Younes M, Lechago LV, Somoano JR, Mosharaf M and Lechago J: Wide expression of the human erythrocyte glucose transporter Glut1 in human cancers. Cancer Res. 56:1164–1167. 1996.PubMed/NCBI

3 

Warburg O: On the origin of cancer cells. Science. 123:309–314. 1956. View Article : Google Scholar : PubMed/NCBI

4 

Plathow C and Weber WA: Tumor cell metabolism imaging. J Nucl Med. 49(Suppl 2): S43–S63. 2008. View Article : Google Scholar

5 

Plas DR and Thompson CB: Akt-dependent transformation: there is more to growth than just surviving. Oncogene. 24:7435–7442. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Elstrom RL, Bauer DE, Buzzai M, et al: Akt stimulates aerobic glycolysis in cancer cells. Cancer Res. 64:3892–3899. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Vousden KH and Ryan KM: p53 and metabolism. Nat Rev Cancer. 9:691–700. 2009. View Article : Google Scholar

8 

Stambolic V, MacPherson D, Sas D, et al: Regulation of PTEN transcription by p53. Mol Cell. 8:317–325. 2001. View Article : Google Scholar : PubMed/NCBI

9 

Jones RG, Plas DR, Kubek S, et al: AMP-activated protein kinase induces a p53-dependent metabolic checkpoint. Mol Cell. 18:283–293. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Shackelford DB and Shaw RJ: The LKB1-AMPK pathway: metabolism and growth control in tumour suppression. Nat Rev Cancer. 9:563–575. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Lu H, Forbes RA and Verma A: Hypoxia-inducible factor 1 activation by aerobic glycolysis implicates the Warburg effect in carcinogenesis. J Biol Chem. 277:23111–23115. 2002. View Article : Google Scholar : PubMed/NCBI

12 

Zhong H, De Marzo AM, Laughner E, et al: Overexpression of hypoxia-inducible factor 1alpha in common human cancers and their metastases. Cancer Res. 59:5830–5835. 1999.PubMed/NCBI

13 

Talks KL, Turley H, Gatter KC, et al: The expression and distribution of the hypoxia-inducible factors HIF-1alpha and HIF-2alpha in normal human tissues, cancers, and tumor-associated macrophages. Am J Pathol. 157:411–421. 2000. View Article : Google Scholar : PubMed/NCBI

14 

Matsuyama T, Nakanishi K, Hayashi T, et al: Expression of hypoxia-inducible factor-1alpha in esophageal squamous cell carcinoma. Cancer Sci. 96:176–182. 2005. View Article : Google Scholar : PubMed/NCBI

15 

Ogane N, Yasuda M, Shimizu M, et al: Clinicopathological implications of expressions of hypoxia-related molecules in esophageal superficial squamous cell carcinoma. Ann Diagn Pathol. 14:23–29. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Zhang ZG, Zhang QN, Wang XH and Tian JH: Hypoxia-inducible factor 1 alpha (HIF-1alpha) as a prognostic indicator in patients with gastric tumors: a meta-analysis. Asian Pac J Cancer Prev. 14:4195–4198. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Lin S, Ma R, Zheng XY, et al: Meta-analysis of immunohistochemical expression of hypoxia inducible factor-1alpha as a prognostic role in gastric cancer. World J Gastroenterol. 20:1107–1113. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Baba Y, Nosho K, Shima K, et al: HIF1A overexpression is associated with poor prognosis in a cohort of 731 colorectal cancers. Am J Pathol. 176:2292–2301. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Zheng SS, Chen XH, Yin X and Zhang BH: Prognostic significance of HIF-1alpha expression in hepatocellular carcinoma: a meta-analysis. PloS One. 8:e657532013. View Article : Google Scholar : PubMed/NCBI

20 

Nakamura J, Kitajima Y, Kai K, et al: Hypoxia-inducible factor-1alpha expression predicts the response to 5-fluorouracil-based adjuvant chemotherapy in advanced gastric cancer. Oncol Rep. 22:693–699. 2009.

21 

Nakamura J, Kitajima Y, Kai K, et al: HIF-1alpha is an unfavorable determinant of relapse in gastric cancer patients who underwent curative surgery followed by adjuvant 5-FU chemotherapy. Int J Cancer. 127:1158–1171. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Griffiths EA, Pritchard SA, McGrath SM, et al: Hypoxia-associated markers in gastric carcinogenesis and HIF-2alpha in gastric and gastro-oesophageal cancer prognosis. Br J Cancer. 98:965–973. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Rasheed S, Harris AL, Tekkis PP, et al: Hypoxia-inducible factor-1alpha and -2alpha are expressed in most rectal cancers but only hypoxia-inducible factor-1alpha is associated with prognosis. Br J Cancer. 100:1666–1673. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Dang CV, Le A and Gao P: MYC-induced cancer cell energy metabolism and therapeutic opportunities. Clin Cancer Res. 15:6479–6483. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Calcagno DQ, Leal MF, Assumpcao PP, Smith MA and Burbano RR: MYC and gastric adenocarcinoma carcinogenesis. World J Gastroenterol. 14:5962–5968. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Liu Y, Gong LP, Dong XL and Liu HG: Detection of C-MYC oncogene translocation and copy number change in the normal-dysplasia- carcinoma sequence of the larynx by fluorescence in situ hybridization. Diagn Cytopathol. 41:515–519. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Tuupanen S, Yan J, Turunen M, et al: Characterization of the colorectal cancer-associated enhancer MYC-335 at 8q24: the role of rs67491583. Cancer Genet. 205:25–33. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Amente S, Lania L and Majello B: Epigenetic reprogramming of Myc target genes. Am J Cancer Res. 1:413–418. 2011.PubMed/NCBI

29 

de Souza CR, Leal MF, Calcagno DQ, et al: MYC deregulation in gastric cancer and its clinicopathological implications. PloS One. 8:e644202013.PubMed/NCBI

30 

He C, Jiang H, Geng S, et al: Expression and prognostic value of c-Myc and Fas (CD95/APO1) in patients with pancreatic cancer. Int J Clin Exp Pathol. 7:742–750. 2014.PubMed/NCBI

31 

Mueckler M, Caruso C, Baldwin SA, et al: Sequence and structure of a human glucose transporter. Science. 229:941–945. 1985. View Article : Google Scholar : PubMed/NCBI

32 

Joost HG and Thorens B: The extended GLUT-family of sugar/polyol transport facilitators: nomenclature, sequence characteristics, and potential function of its novel members (review). Mol Membr Biol. 18:247–256. 2001. View Article : Google Scholar

33 

Younes M, Lechago LV, Somoano JR, Mosharaf M and Lechago J: Immunohistochemical detection of Glut3 in human tumors and normal tissues. Anticancer Res. 17:2747–2750. 1997.PubMed/NCBI

34 

Ayala FR, Rocha RM, Carvalho KC, et al: GLUT1 and GLUT3 as potential prognostic markers for oral squamous cell carcinoma. Molecules. 15:2374–2387. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Fonteyne P, Casneuf V, Pauwels P, et al: Expression of hexokinases and glucose transporters in treated and untreated oesophageal adenocarcinoma. Histol Histopathol. 24:971–977. 2009.PubMed/NCBI

36 

Carvalho KC, Cunha IW, Rocha RM, et al: GLUT1 expression in malignant tumors and its use as an immunodiagnostic marker. Clinics (Sao Paulo). 66:965–972. 2011. View Article : Google Scholar : PubMed/NCBI

37 

Griffiths EA, Pritchard SA, Welch IM, Price PM and West CM: Is the hypoxia-inducible factor pathway important in gastric cancer? Eur J Cancer. 41:2792–2805. 2005. View Article : Google Scholar : PubMed/NCBI

38 

Yun J, Rago C, Cheong I, et al: Glucose deprivation contributes to the development of KRAS pathway mutations in tumor cells. Science. 325:1555–1559. 2009. View Article : Google Scholar : PubMed/NCBI

39 

Kawada K, Nakamoto Y, Kawada M, et al: Relationship between 18F-fluorodeoxyglucose accumulation and KRAS/BRAF mutations in colorectal cancer. Clin Cancer Res. 18:1696–1703. 2012.

40 

Yang W, Zheng Y, Xia Y, et al: ERK1/2-dependent phosphorylation and nuclear translocation of PKM2 promotes the Warburg effect. Nat Cell Biol. 14:1295–1304. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Young CD, Lewis AS, Rudolph MC, et al: Modulation of glucose transporter 1 (GLUT1) expression levels alters mouse mammary tumor cell growth in vitro and in vivo. PloS One. 6:e232052011. View Article : Google Scholar : PubMed/NCBI

42 

Sawayama H, Ishimoto T, Watanabe M, et al: High expression of glucose transporter 1 on primary lesions of esophageal squamous cell carcinoma is associated with hematogenous recurrence. Ann Surg Oncol. 21:1756–1762. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Tohma T, Okazumi S, Makino H, et al: Overexpression of glucose transporter 1 in esophageal squamous cell carcinomas: a marker for poor prognosis. Dis Esophagus. 18:185–189. 2005. View Article : Google Scholar : PubMed/NCBI

44 

Kawamura T, Kusakabe T, Sugino T, et al: Expression of glucose transporter-1 in human gastric carcinoma: association with tumor aggressiveness, metastasis, and patient survival. Cancer. 92:634–641. 2001. View Article : Google Scholar

45 

Jung JH, Im S, Jung ES and Kang CS: Clinicopathological implications of the expression of hypoxia-related proteins in gastric cancer. Int J Med Sci. 10:1217–1223. 2013. View Article : Google Scholar : PubMed/NCBI

46 

Haber RS, Rathan A, Weiser KR, et al: GLUT1 glucose transporter expression in colorectal carcinoma: a marker for poor prognosis. Cancer. 83:34–40. 1998. View Article : Google Scholar : PubMed/NCBI

47 

Korkeila E, Jaakkola PM, Syrjanen K, Pyrhonen S and Sundstrom J: Pronounced tumour regression after radiotherapy is associated with negative/weak glucose transporter-1 expression in rectal cancer. Anticancer Res. 31:311–315. 2011.PubMed/NCBI

48 

Kitamura K, Hatano E, Higashi T, et al: Proliferative activity in hepatocellular carcinoma is closely correlated with glucose metabolism but not angiogenesis. J Hepatol. 55:846–857. 2011. View Article : Google Scholar : PubMed/NCBI

49 

Legan M, Tevzic S, Tolar A, Luzar B and Marolt VF: Glucose transporter-1 (GLUT-1) immunoreactivity in benign, premalignant and malignant lesions of the gallbladder. Pathol Oncol Res. 17:61–66. 2011. View Article : Google Scholar : PubMed/NCBI

50 

Kim YW, Park YK, Yoon TY and Lee SM: Expression of the GLUT1 glucose transporter in gallbladder carcinomas. Hepatogastroenterology. 49:907–911. 2002.PubMed/NCBI

51 

Sattler UG and Mueller-Klieser W: The anti-oxidant capacity of tumour glycolysis. Int J Radiat Biol. 85:963–971. 2009. View Article : Google Scholar : PubMed/NCBI

52 

Hirschhaeuser F, Sattler UG and Mueller-Klieser W: Lactate: a metabolic key player in cancer. Cancer Res. 71:6921–6925. 2011. View Article : Google Scholar : PubMed/NCBI

53 

Brophy S, Sheehan KM, McNamara DA, Deasy J, Bouchier-Hayes DJ and Kay EW: GLUT-1 expression and response to chemoradiotherapy in rectal cancer. Int J Cancer. 125:2778–2782. 2009. View Article : Google Scholar : PubMed/NCBI

54 

Cao X, Fang L, Gibbs S, et al: Glucose uptake inhibitor sensitizes cancer cells to daunorubicin and overcomes drug resistance in hypoxia. Cancer Chemother Pharmacol. 59:495–505. 2007. View Article : Google Scholar : PubMed/NCBI

55 

Wu CH, Ho YS, Tsai CY, et al: In vitro and in vivo study of phloretin-induced apoptosis in human liver cancer cells involving inhibition of type II glucose transporter. Int J Cancer. 124:2210–2219. 2009. View Article : Google Scholar : PubMed/NCBI

56 

Liu Y, Cao Y, Zhang W, et al: A small-molecule inhibitor of glucose transporter 1 downregulates glycolysis, induces cell-cycle arrest, and inhibits cancer cell growth in vitro and in vivo. Mol Cancer Ther. 11:1672–1682. 2012. View Article : Google Scholar : PubMed/NCBI

57 

Mathupala SP, Ko YH and Pedersen PL: Hexokinase-2 bound to mitochondria: cancer’s stygian link to the ‘Warburg Effect’ and a pivotal target for effective therapy. Semin Cancer Biol. 19:17–24. 2009.

58 

Kwee SA, Hernandez B, Chan O and Wong L: Choline kinase alpha and hexokinase-2 protein expression in hepatocellular carcinoma: association with survival. PloS One. 7:e465912012. View Article : Google Scholar : PubMed/NCBI

59 

Paudyal B, Paudyal P, Oriuchi N, Tsushima Y, Nakajima T and Endo K: Clinical implication of glucose transport and metabolism evaluated by 18F-FDG PET in hepatocellular carcinoma. Int J Oncol. 33:1047–1054. 2008.PubMed/NCBI

60 

Seo S, Hatano E, Higashi T, et al: Fluorine-18 fluorodeoxyglucose positron emission tomography predicts tumor differentiation, P-glycoprotein expression, and outcome after resection in hepatocellular carcinoma. Clin Cancer Res. 13:427–433. 2007. View Article : Google Scholar

61 

Ganapathy-Kanniappan S, Vali M, Kunjithapatham R, et al: 3-bromopyruvate: a new targeted antiglycolytic agent and a promise for cancer therapy. Curr Pharm Biotechnol. 11:510–517. 2010. View Article : Google Scholar : PubMed/NCBI

62 

David CJ, Chen M, Assanah M, Canoll P and Manley JL: HnRNP proteins controlled by c-Myc deregulate pyruvate kinase mRNA splicing in cancer. Nature. 463:364–368. 2010. View Article : Google Scholar : PubMed/NCBI

63 

Clower CV, Chatterjee D, Wang Z, Cantley LC, Vander Heiden MG and Krainer AR: The alternative splicing repressors hnRNP A1/A2 and PTB influence pyruvate kinase isoform expression and cell metabolism. Proc Natl Acad Sci USA. 107:1894–1899. 2010. View Article : Google Scholar

64 

Christofk HR, Vander Heiden MG, Harris MH, et al: The M2 splice isoform of pyruvate kinase is important for cancer metabolism and tumour growth. Nature. 452:230–233. 2008. View Article : Google Scholar : PubMed/NCBI

65 

Christofk HR, Vander Heiden MG, Wu N, Asara JM and Cantley LC: Pyruvate kinase M2 is a phosphotyrosine-binding protein. Nature. 452:181–186. 2008. View Article : Google Scholar : PubMed/NCBI

66 

Anastasiou D, Yu Y, Israelsen WJ, et al: Pyruvate kinase M2 activators promote tetramer formation and suppress tumorigenesis. Nat Chem Biol. 8:839–847. 2012. View Article : Google Scholar : PubMed/NCBI

67 

Yang W and Lu Z: Regulation and function of pyruvate kinase M2 in cancer. Cancer Lett. 339:153–158. 2013. View Article : Google Scholar : PubMed/NCBI

68 

Tamada M, Suematsu M and Saya H: Pyruvate kinase M2: multiple faces for conferring benefits on cancer cells. Clin Cancer Res. 18:5554–5561. 2012. View Article : Google Scholar : PubMed/NCBI

69 

Kaplon J, Zheng L, Meissl K, et al: A key role for mitochondrial gatekeeper pyruvate dehydrogenase in oncogene-induced senescence. Nature. 498:109–112. 2013. View Article : Google Scholar : PubMed/NCBI

70 

Hur H, Xuan Y, Kim YB, et al: Expression of pyruvate dehydrogenase kinase-1 in gastric cancer as a potential therapeutic target. Int J Oncol. 42:44–54. 2013.PubMed/NCBI

71 

Lu CW, Lin SC, Chien CW, et al: Overexpression of pyruvate dehydrogenase kinase 3 increases drug resistance and early recurrence in colon cancer. Am J Pathol. 179:1405–1414. 2011. View Article : Google Scholar : PubMed/NCBI

72 

Peifer C and Alessi DR: Small-molecule inhibitors of PDK1. Chem Med Chem. 3:1810–1838. 2008. View Article : Google Scholar : PubMed/NCBI

73 

Tong J, Xie G, He J, Li J, Pan F and Liang H: Synergistic antitumor effect of dichloroacetate in combination with 5-fluorouracil in colorectal cancer. J Biomed Biotechnol. 2011:7405642011. View Article : Google Scholar : PubMed/NCBI

74 

Shen YC, Ou DL, Hsu C, et al: Activating oxidative phosphorylation by a pyruvate dehydrogenase kinase inhibitor overcomes sorafenib resistance of hepatocellular carcinoma. Br J Cancer. 108:72–81. 2013. View Article : Google Scholar

75 

Fantin VR, St-Pierre J and Leder P: Attenuation of LDH-A expression uncovers a link between glycolysis, mitochondrial physiology, and tumor maintenance. Cancer Cell. 9:425–434. 2006. View Article : Google Scholar : PubMed/NCBI

76 

Goldman RD, Kaplan NO and Hall TC: Lactic dehydrogenase in human neoplastic tissues. Cancer Res. 24:389–399. 1964.PubMed/NCBI

77 

Fan J, Hitosugi T, Chung TW, et al: Tyrosine phosphorylation of lactate dehydrogenase A is important for NADH/NAD(+) redox homeostasis in cancer cells. Mol Cell Biol. 31:4938–4950. 2011. View Article : Google Scholar : PubMed/NCBI

78 

Koukourakis MI, Giatromanolaki A, Sivridis E, Gatter KC and Harris AL: Lactate dehydrogenase 5 expression in operable colorectal cancer: strong association with survival and activated vascular endothelial growth factor pathway - a report of the Tumour Angiogenesis Research Group. J Clin Oncol. 24:4301–4308. 2006. View Article : Google Scholar

79 

Sheng SL, Liu JJ, Dai YH, Sun XG, Xiong XP and Huang G: Knockdown of lactate dehydrogenase A suppresses tumor growth and metastasis of human hepatocellular carcinoma. FEBS J. 279:3898–3910. 2012. View Article : Google Scholar

80 

Zhang Y, Zhang X, Wang X, et al: Inhibition of LDH-A by lentivirus-mediated small interfering RNA suppresses intestinaltype gastric cancer tumorigenicity through the downregulation of Oct4. Cancer Lett. 321:45–54. 2012. View Article : Google Scholar : PubMed/NCBI

81 

Le A, Cooper CR, Gouw AM, et al: Inhibition of lactate dehydrogenase A induces oxidative stress and inhibits tumor progression. Proc Natl Acad Sci USA. 107:2037–2042. 2010. View Article : Google Scholar : PubMed/NCBI

82 

Miskimins WK, Ahn HJ, Kim JY, Ryu S, Jung YS and Choi JY: Synergistic anti-cancer effect of phenformin and oxamate. PloS One. 9:e855762014. View Article : Google Scholar : PubMed/NCBI

83 

Zhou M, Zhao Y, Ding Y, et al: Warburg effect in chemosensitivity: targeting lactate dehydrogenase-A re-sensitizes taxol-resistant cancer cells to taxol. Mol Cancer. 9:332010. View Article : Google Scholar : PubMed/NCBI

84 

Sumiyoshi Y, Kakeji Y, Egashira A, Mizokami K, Orita H and Maehara Y: Overexpression of hypoxia-inducible factor 1alpha and p53 is a marker for an unfavorable prognosis in gastric cancer. Clin Cancer Res. 12:5112–5117. 2006. View Article : Google Scholar : PubMed/NCBI

85 

Isobe T, Aoyagi K, Koufuji K, et al: Clinicopathological significance of hypoxia-inducible factor-1 alpha (HIF-1alpha) expression in gastric cancer. Int J Clin Oncol. 18:293–304. 2013. View Article : Google Scholar : PubMed/NCBI

86 

Theodoropoulos GE, Lazaris AC, Theodoropoulos VE, et al: Hypoxia, angiogenesis and apoptosis markers in locally advanced rectal cancer. Int J Colorectal Dis. 21:248–257. 2006. View Article : Google Scholar : PubMed/NCBI

87 

Dai CX, Gao Q, Qiu SJ, et al: Hypoxia-inducible factor-1 alpha, in association with inflammation, angiogenesis and MYC, is a critical prognostic factor in patients with HCC after surgery. BMC Cancer. 9:4182009. View Article : Google Scholar : PubMed/NCBI

88 

Liu L, Zhu XD, Wang WQ, et al: Activation of beta-catenin by hypoxia in hepatocellular carcinoma contributes to enhanced metastatic potential and poor prognosis. Clin Cancer Res. 16:2740–2750. 2010. View Article : Google Scholar : PubMed/NCBI

89 

Couvelard A, O’Toole D, Leek R, et al: Expression of hypoxia-inducible factors is correlated with the presence of a fibrotic focus and angiogenesis in pancreatic ductal adenocarcinomas. Histopathology. 46:668–676. 2005. View Article : Google Scholar : PubMed/NCBI

90 

Shen YM, Arbman G, Olsson B and Sun XF: Overexpression of GLUT1 in colorectal cancer is independently associated with poor prognosis. Int J Biol Markers. 26:166–172. 2011. View Article : Google Scholar : PubMed/NCBI

91 

Pizzi S, Porzionato A, Pasquali C, et al: Glucose transporter-1 expression and prognostic significance in pancreatic carcinogenesis. Histol Histopathol. 24:175–185. 2009.PubMed/NCBI

92 

Rho M, Kim J, Jee CD, et al: Expression of type 2 hexokinase and mitochondria-related genes in gastric carcinoma tissues and cell lines. Anticancer Res. 27:251–258. 2007.PubMed/NCBI

93 

Qiu MZ, Han B, Luo HY, et al: Expressions of hypoxia-inducible factor-1alpha and hexokinase-II in gastric adenocarcinoma: the impact on prognosis and correlation to clinicopathologic features. Tumour Biol. 32:159–166. 2011. View Article : Google Scholar : PubMed/NCBI

94 

Gong L, Cui Z, Chen P, Han H, Peng J and Leng X: Reduced survival of patients with hepatocellular carcinoma expressing hexokinase II. Med Oncol. 29:909–914. 2012. View Article : Google Scholar

95 

Zhan C, Shi Y, Lu C and Wang Q: Pyruvate kinase M2 is highly correlated with the differentiation and the prognosis of esophageal squamous cell cancer. Dis Esophagus. 26:746–753. 2013.PubMed/NCBI

96 

Lim JY, Yoon SO, Seol SY, et al: Overexpression of the M2 isoform of pyruvate kinase is an adverse prognostic factor for signet ring cell gastric cancer. World J Gastroenterol. 18:4037–4043. 2012. View Article : Google Scholar : PubMed/NCBI

97 

Li J, Yang Z, Zou Q, et al: PKM2 and ACVR 1C are prognostic markers for poor prognosis of gallbladder cancer. Clin Transl Oncol. 16:200–207. 2014. View Article : Google Scholar : PubMed/NCBI

98 

Kolev Y, Uetake H, Takagi Y and Sugihara K: Lactate dehydrogenase- 5 (LDH-5) expression in human gastric cancer: association with hypoxia-inducible factor (HIF-1alpha) pathway, angiogenic factors production and poor prognosis. Ann Surg Oncol. 15:2336–2344. 2008. View Article : Google Scholar

99 

Yu SJ, Yoon JH, Yang JI, et al: Enhancement of hexokinase II inhibitor-induced apoptosis in hepatocellular carcinoma cells via augmenting ER stress and anti-angiogenesis by protein disulfide isomerase inhibition. J Bioenerg Biomembr. 44:101–115. 2012. View Article : Google Scholar

100 

Zhou Y, Tozzi F, Chen J, et al: Intracellular ATP levels are a pivotal determinant of chemoresistance in colon cancer cells. Cancer Res. 72:304–314. 2012. View Article : Google Scholar : PubMed/NCBI

101 

Vander Heiden MG, Christofk HR, Schuman E, et al: Identification of small molecule inhibitors of pyruvate kinase M2. Biochem Pharmacol. 79:1118–1124. 2010.PubMed/NCBI

102 

Feldman RI, Wu JM, Polokoff MA, et al: Novel small molecule inhibitors of 3-phosphoinositide-dependent kinase-1. J Biol Chem. 280:19867–19874. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October 2014
Volume 45 Issue 4

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Sawayama H, Ishimoto T, Sugihara H, Miyanari N, Miyamoto Y, Baba Y, Yoshida N and Baba H: Clinical impact of the Warburg effect in gastrointestinal cancer (Review). Int J Oncol 45: 1345-1354, 2014
APA
Sawayama, H., Ishimoto, T., Sugihara, H., Miyanari, N., Miyamoto, Y., Baba, Y. ... Baba, H. (2014). Clinical impact of the Warburg effect in gastrointestinal cancer (Review). International Journal of Oncology, 45, 1345-1354. https://doi.org/10.3892/ijo.2014.2563
MLA
Sawayama, H., Ishimoto, T., Sugihara, H., Miyanari, N., Miyamoto, Y., Baba, Y., Yoshida, N., Baba, H."Clinical impact of the Warburg effect in gastrointestinal cancer (Review)". International Journal of Oncology 45.4 (2014): 1345-1354.
Chicago
Sawayama, H., Ishimoto, T., Sugihara, H., Miyanari, N., Miyamoto, Y., Baba, Y., Yoshida, N., Baba, H."Clinical impact of the Warburg effect in gastrointestinal cancer (Review)". International Journal of Oncology 45, no. 4 (2014): 1345-1354. https://doi.org/10.3892/ijo.2014.2563