Gastric cancer: Metabolic and metabolomics perspectives (Review)

  • Authors:
    • Shiyu Xiao
    • Liya Zhou
  • View Affiliations

  • Published online on: May 16, 2017     https://doi.org/10.3892/ijo.2017.4000
  • Pages: 5-17
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Gastric cancer is one of the most malignant tumors worldwide and remains a major health threat in Asia-Pacific regions, while its pathological mechanism is generally unknown. Recent research has advanced the understanding of the relationship between metabolic reprogramming and carcinogenesis. In particular, metabolic regulation and cancer research are being further brought into sharp focus with the emergence of metabolomics. Not only can metabolomics provide global information on metabolic profiles of specific tumors, but it can also act as a promising tool to discover biomarkers regarding diagnosis, metastatic surveillance and chemotherapeutic sensitivity prediction. Meanwhile, metabolism-based anticancer therapies will be further discovered. Up to now, accumulative studies have highlighted the application of metabolomics in gastric cancer research regarding different aspects; therefore we summarized the current available results of how metabolic changes are linked to gastric carcinogenesis, and how metabolomics holds promise for the diagnosis, metastatic surveillance, treatment and prognosis prediction of gastric cancer.

1. Introduction

Gastric cancer remains third in ranking in cancer death worldwide, although its overall incidence is declining in recent years (1). In the past decades, studies aimed at Helicobacter pylori infection (2,3), hereditary susceptibility (4) and environmental factors (5) have made a great breakthrough in investigating its precise pathogenesis. Recently, application of various '-omics' technologies opened a new field to investigate the mechanisms behind this disease.

With the emergency of metabolomics, major progress has been made in the understanding of the relationship between metabolic regulation and cancer. Warburg, in fact, showed a characteristic metabolic pattern of tumors in the 1920s, that is, tumor cells consume a large amount of glucose for glycolysis even under the condition of sufficient oxygen (Warburg effect) (6). Extensive research also indicates that metabolic reprogramming is one of the hallmarks of cancer (7), and intricately linked to oncogenesis (810) and cancer immune escape (1113). On the other hand, study methods combined conventional oncology research and metabolomics are more likely to provide deeper insights in this field. The procedure of these methods is illustrated in Fig. 1, and more detailed information can be found in literature (1416).

Several excellent reviews have been published on metabolomics application in different diseases (1719) especially cancer research (2023). Hence, this report presents fresh and profound insights into metabolic changes in gastric cancer and possible mechanism behind these alterations is further discussed. Then, we focus on some studies including our data targeted on biomarkers involving diagnosis, metastasis and prognosis, and treatment in this disease. Finally, future directions are presented.

2. Metabolic alteration in gastric cancer

Up to now, several studies aimed at identifiable metabolic changes in macroenvironment-blood (2429) (Table I) and urine (3034) (Table II) or microenvironment-carcinoma tissues (3541) (Table III) and gastric juice (4244) (Table IV) have been done to map globally metabolic profiles and interpret its possible mechanism in the process of gastric carcinogenesis. Typical changes in metabolites of this disease are illustrated in Fig. 2.

Table I

Metabolites in blood samples between cancer groups and non-cancer groups.

Table I

Metabolites in blood samples between cancer groups and non-cancer groups.

YearPatients/animal modelsSamplesSample sizeMethodMajor findingsRef.
2011PatientsPlasmaHealthy people (n=985)
Gastric cancer (n=199)
HPLC-ESI-MSi) Concentration of amino acids was significantly decreased in plasma of gastric cancer patients and this change occurs in early stage regardless of the subsequent progression and poor nutrition
ii) Plasma-free amino acids have shared alteration among gastric cancer, lung cancer, colorectal cancer, breast cancer and prostate cancer, but specific profiles in gastric cancer were also detected
(24)
2011PatientsPlasmaChronic superficial gastritis (n=19)
Chronic atrophic gastritis (n=10)
Intestinal metaplasia (n=10)
Dysplasia (n=15)
Gastric cancer (n=22)
GC-TOFMSi) 15 metabolites (increase, glutamate, asparagine, ornithine, pyroglutamate, 2-hydroxybutyrate, azelaic acid, 11-eicosenoic acid, 1-monohexadecanoylglycerol-γ-tocopherol, urate; decrease: creatinine, threonate) were different between chronic superficial gastritis and gastric cancer group
ii) Metabolic phenotype of gastric cancer was greatly similar to intestinal metaplasia
(25)
2012PatientsSerumHealthy people (n=30)
Gastric cancer (n=30)
GC-MSi) 18 metabolites were different between gastric cancer and healthy control group, including fumaric acid, glutamine, valine, sarcosine, 9,12-octadecadienoic acid, 9-octadecenoic acid, trans-13-octadecenoic acid, nonahexacontanoic, cholesta-3,5-diene, cholesterol pentafluoropionate, cholesterol, Cholest-5-en-3-ol, 2-O-mesyl arabinose, hexadecanenitrile, benzeneacetonitrile, 2-amino-4-hydroxy-pteridinone, 1,2,4-benzenetricarboxylic acid and hexanedioic acid
ii) Valine showed the greatest fold change and hexanedioic acid was the most depleted
(26)
2012PatientsSerumHealthy people (n=12)
Gastric cancer (n=11)
GC-MSi) Pyruvic acid, 3-hydroxypropionic acid, 3-hydroxyisobutyric acid, octanoic acid, phosphoric acid significantly changed in gastric cancer patients
ii) Levels of 3-hydroxypropionic acid and pyruvic acid could discriminate gastric cancer from esophageal or colorectal cancer
(27)
2012PatientsPlasmaChronic superficial gastritis (n=20)
Gastric cancer (n=17)
Post-operation of gastric cancer (n=15)
GC-TOFMSi) 15 discriminatory metabolites (increase, β-hydroxybutyrate, β-D-methylglucopyranoside, heptanoic acid; decrease, succinate, malate, fumarate, citrate, serine, glycine, cysteine, and S-methyl-cysteine, docosahexaenoic acid, inositol-phosphate, octadecenoic acid, and 9-(Z)-hexadecenoic acid) were identified between gastric cancer and chronic superficial gastritis group
ii) Surgical removal of the cancer tissues could change levels of many metabolites that characterized the metabolic phenotype of the cancer group
(28)
2016PatientsSerumChronic superficial gastritis (n=17)
Gastric cancer (n=32)
LC-MSGastric cancer displays upregulated kynurenine pathway of tryptophan metabolism: increase, indole-3-lactic acid, anthranilic acid, kynurenic acid; decrease, kynurenine, 3-indoxyl-sulfate(29)

[i] HPLC-ESI-MS, high performance liquid chromatography-electrospray ionization-mass spectrometry; GC-TOMS, gas chromatography-time-of-flight mass spectrometry; GC-MS, gas chromatography-mass spectrometry; LC-MS, liquid chromatography-mass spectrometry.

Table II

Metabolites in urine samples between cancer groups and non-cancer groups.

Table II

Metabolites in urine samples between cancer groups and non-cancer groups.

YearPatients/animal modelsSample sizeMethodMajor findingsRef
2011SCID mice (male) SCG-7901 cell lineGastric cancer (n=16) (metastasis group (=8 and non-metastasis (=8)
Control group (n=8)
GC-MSi) 10 metabolites were different between cancer group (metastasis and non-metastasis) and control group: lactic acid, malic acid, citric acid, glycerol, hexadecanoic acid, pyrimidine, uric acid, butanoic acid, propanoic, butanedioic acid
ii) 7 metabolites were characteristic between metastasis and non-metastasis groups: alanine, L-proline, glycerol, butanoic acid, butanedioic acid, L-threonic acid, myo-inositol. iii) Changes in lactic acid and butanoic acid showed diagnostic value
(30)
2014PatientsTraining set/validation set
Gastric cancer (n=50/23)
Healthy people (n=50/31)
1H-NMRi) Altered metabolites in urine samples of gastric cancer are mainly related to amino acids and lipid metabolism; levels of 4-hydroxyphenylacetate, alamine, phenylacetylglycine, manmitol, glycolate and arginine are related to T stage of gastric cancer
ii) Hypoxanthine is able to predict a recovery trend in postoperative groups of gastric cancer
(31)
2015PatientsGastric cancer (n=13)
Healthy people (n=9)
LC-MS16 metabolites were differently expressed between cancer and healthy group: succinic acid, malic acid, alanine, glycine, L-proline, hexadecanoic acid, pyrimidine, uric acid, glycocholic acid, hippurate, urea, 4-deoxythreonic acid, phenylacetylglycine, taurine, 2-oxoglutarate(32)
2016PatientsGastric cancer (n=43)
Healthy people (n=40)
Barrett's esophagus (n=40)
1H-NMRi) Levels of sucrose, dimethylamine, 1-methylnicotinamide, 2-furoylgylycine, N-acetyl-serotonin, trans-aconitate, formate and serotonin greatly altered in urine samples of gastric cancer
ii) Alanine, 2-hydroxyisobutyrate and 3-indoxylsulfate produced a discriminatory model regarding discriminating cancer and control group
(33)
2016PatientsGastric cancer (n=199)
Healthy people (n=87)
GC-MSi) 17 metabolites are largely different between cancer and control groups in training set: glycine, valine, isoleucine, serine, threonine, proline, methionine, tyrosine, tryptophan, ethyl 2-methylacetoacetate, levulinic acid, p-cresol, benzylmalonic acid, 4-hydroxybenzoic acid, hippuric acid, benzil, alamine
ii) 14 of them show diagnostic value that is better than classic blood biomarkers on validation set, most of which were related to amino acid metabolism
iii) Proline, p-cresol and 4-hydroxybenzoic acid produce outcome-prediction value by survival analysis
(34)

[i] SCID, severe combined immune deficiency; 1H-NMR, 1hydrogen-nuclear magnetic resonance; LC-MS, liquid chromatography-mass spectrometry.

Table III

Metabolites in tissue samples between cancer groups and non-cancer groups.

Table III

Metabolites in tissue samples between cancer groups and non-cancer groups.

YearPatients/animal modelsSample sizeMethodMajor findingsRef.
2009Patients12 pairs of matched tumor and normal gastric tissuesCE-TOFMSi) Extremely low glucose, high lactate and glycolytic intermediate concentrations were found in both colon and stomach tumor tissues
ii) Significant accumulation of all amino acids except glutamine in the tumor tissues
(35)
2010Male SCID mice
SCG-7901 cell line
Cancer group
(Non-metastasis 8 Metastasis 8)
Control group 6
GC-MSi) 29 metabolites were differently expressed between metastasis and non-metastasis group: glucose, succinate, malicacid, lactate, alanine, glycine, valine, leucine, dimethylglycine, isoleucine, propanamide, butanedioic, proline, methionnine, serine, threonine, asparagine, glutamine, phosphoserine, glutamate, lysine, arginine, docosanoic, octadecanoic, pyrimidine, hypoxanthine, inositol, propanedioc, pyrrolidine
ii) Serine and proline metabolisms were highlighted in metastatic group
(36)
2010Patients18 pairs of matched tumor and normal gastric tissuesGC-MSi) L-glutamine, phosphoserine, L-valine, L-isoleucine, serine, heptanedioic acid, propanoic acid, phenanthrenol, butanetriol, acetamid, butenoic acid, oxazolethione, naphthalene, L-altrose, L-mannofuranose, galactofuranoside, myo-inositol, D-ribofuranose were detected differently between the malignant tissues and the adjacent non-malignant tissues of gastric mucosa
ii) 5 of them were detected differently between the non-invasive tumors and the invasive tumors: higher levels of L-cysteine, L-tyrosine, hypoxanthine and lower levels of phenanthrenol, butanoic acid in the invasive group
(37)
2010Patients65 pairs of matched gastric cardiac cancer and adjacent normal tissuesGC-TOFMSi) Dysregulation of pyruvic acid efflux was an important glucose metabolic signature in the development of gastric cardiac cancer
ii) Transition from glycolysis to the Krebs cycle had an inhibitory effect on GCC progression, which could be served as a potential therapeutic target for gastric cardiac cancer
(38)
2011Patients30 pairs of matched tumor and normal gastric tissuesGC-MSi) 15 differential metabolites were identified: α-ketoglutaric acid, fumaric acid, valric acid, 9-hexadecennoic acid, 3-hydroxybutanoic acid, hexadecanoic acid, octadecanoic acid, cis-vaccenic acid, arachidonic acid, 1-phenanthrene-carboxylic acid, 9-octadecenamide, squalene, xylonic acid, benzenepropanoic acid
ii) Current models could not discriminate normal mucosa and different pathological stages of GC tissues based on their identified metabolic profiles
(39)
2012PatientsGastric cancer (n=17)
Chronic superficial gastritis (n=20)
GC-TOFMSDiscriminating metabolites associated with glucose, amino acids, lipid and nucleotide metabolism were detected between two groups: increase, citrate, malate, fumarate, succinate), cysteine, 2-aminoadipate, 9-(Z)-hexadecenoic acid, docosahexaenoic acid, β-hydroxybutyrate, uracil, monomethylphosphate; decrease, glucose, maltose, ribose, β-D-methylglucopyranoside, fructose-6-phosphate, inositol and ribitol, glyceric acid-2,3-diphosphate, nonesterified cholesterol, uridine(28)
(31)
2014Patients30 pairs of matched tumor and normal gastric tissuesHR-MAS-NMRLipid metabolites were significantly lower, while some amino acids (such as isoleucine, glutamate, leucine, valine, alanine, lysine and phenylalanine), taurine and lactate were significantly higher in tumor tissues

[i] CE-TOFMS, capillary electrophoresis time-of-flight mass spectrometry; HR-MAS NMR, high-resolution magic angle spinning nuclear magnetic resonance.

Table IV

Metabolites in gastric juice between cancer groups and non-cancer groups.

Table IV

Metabolites in gastric juice between cancer groups and non-cancer groups.

YearPatients/animal modelsSample sizeMethodMajor findingsRef.
2011PatientsBenign gastric diseases (n=68)
Gastric malignancies (n=33)
HPLC
LC-MS
i) Aromatic amino acids in gastric juice can be used as diagnostic biomarkers to screen gastric malignancies, areas under receiver operating characteristic curves for tyrosine, phenylalanine and tryptophan were 0.838, 0.856 and 0.816, respectively
ii) The sensitivity and specificity of gastric malignancy detection with phenylalanine reached 87.9% and 79.4%
(40)
2012PatientsNon-neoplastic gastric disease (n=70)
Early gastric cancer (n=49)
Advanced gastric cancer (n=66)
HPLCLevels of tyrosine, phenylalanine and tryptophan in gastric juice increased in the early phase of gastric carcinogenesis, which could function as a biomarker to screen this disease at early stage in the general population(41)
2016PatientsChronic superficial gastritis (n=17)
Gastric cancer (n=32)
LC-MSUpregulated kynurenine pathway of tryptophan metabolism: levels of tryptophan, anthranilic acid, nicotinic acid, kynurenic acid, kynurenine and indole-3-lactic acid (P>0.05) were increased(29)

[i] HPLC, high performance liquid chromatography.

Glucose metabolism

Cumulative evidence demonstrates that concentration of lactic acid shows a consistent increase in urine (30) or tissue (31,35,36,38) samples of gastric cancer groups, but glucose is considerably depleted compared with those healthy counterparts or non-malignant patients (like chronic superficial gastritis and chronic atrophic gastritis without intestinal metaplasia) (35,36). The high lactate level might be attributed to the special metabolism of most cancer cells, known as 'Warburg effect' we mentioned above (6). Scarce glucose might result from the overexpression of glucose transporters (42) and type II hexokinase (43), which are both confirmed in gastric cancer tissues. Higher fructose-6-phosphokinase (6-FPK) activity can also result in low glucose in gastric cancer tissues (44), as it regulates the output of glucose to glycolysis pathway. The glycolytic switch has been identified to be associated with oncogenic transformation and molecular signal transduction, such as hypoxia-inducible factor pathway, insulin signaling pathway and PI3K-Akt-mTOR pathway (45). Furthermore, overexpression of pyruvate kinase and lactate dehydrogenase is positively associated with tumor proliferation and poor prognosis, downregulation of them in vitro experiment can impair tumor invasion (38,4649). On the other hand, such special microenvironment might be the requirement of rapid propagation of tumor cells. To our understanding, it has been reported that accumulated lactic acid moderates the activity of proteases that decompose extracellular matrix, which can produce some peptides and amino acids that are consumable for energy generation (44). Acidosis microenvironment is also ascribed to the formation of cancer blood vessels, meeting the plentiful supply of nutrients and leading to tumor invasion and metastasis (50). Moreover, tumor-derived lactate shows strongly negative effects on cytotoxic T-cell/NK cell function (11,51) and blocks differentiation of monocytes to dendritic cells (52), finally leading to tumor immune escape. However, such outcome demands further verification in gastric cancer.

Considering tricarboxylic acid cycle (TCA) intermediates, an increase of five metabolites (α-ketoglutaric acid, malic acid, fumarate, succinate, citric acid) is noticed regardless of blood (26,28), urine (30,32) or tissue (28,35,36,39) samples in gastric cancer. There are some possible reasons that can explain this phenomenon. One account is that cancer cells still use a small portion of glucose for oxidative phosphorylation. Secondly, cancer cells might also utilize fumarate respiration to generate energy under special conditions of glucose deprivation and severe hypoxia in microenvironment (53), and succinate is one of the byproducts in this process except for originating from TCA. Hence, it provides a likely explanation for the accumulation of fumarate and succinate. Another reason is that some amino acids, such as glutamine, threonine, phenylalanine, tyrosine or proline, can be converted into these intermediates involving in TCA (Fig. 2). Additionally, elevated levels of citric acid can be used in the de novo fatty acid synthesis, but it is noted that citrate can also induce apoptosis in two gastric cancer cell lines in vitro experiment (54,55).

Amino acid metabolism

Availability of amino acids is pivotal for cellular protein biosynthesis and cytoskeleton formation, while it has been pointed out that amino acids especially those linking to TCA (Fig. 2) are an alternative energy source of cancer cell proliferation (56). By employing metabolomics technologies, levels of various amino acids (including serine, valine, phenylalanine, tryptophan, glycine, and proline) and their primary derivatives (such as kynurenine, kynurenic acid, anthranilic acid and nicotinic acid) are significantly higher in tissue specimens (31,36,37) and gastric content (29,40,41), but decreased concentration in some of them is observed in blood (24). The overexpression of L-type amino acid transporter 1 (LAT1) might be proposed to explain this dissimilarity (57). Free amino acids are greatly assimilated to cancer tissues via LAT1 from bloodstream, resulting in the low accumulation of amino acid in contrast to normal counterparts. Malnourishment may also be a contributing factor to these reduced levels of plasma amino acids. Apart from these, degradation of extracellular matrix mediated by the overexpressed matrix metalloproteinases (MMPs) and activated autophagic degradation of intracellular proteins are considered as the potential source of accumulative amino acids in tumor tissues (5860).

Elevated amino acids in microenvironment are contributing factors in carcinogenesis. Most strikingly, it is indicated that many cancer cell lines cannot survive in the absence of glutamine (61), because it is required for anabolic growth of mammalian cells through its ability to control the master regulator of protein translation mTORC1 (62). Reprogramming of glutamine metabolism further contributes to the proliferative and metabolic responses regulated by oncogenic transcription factor c-MYC (63). In addition, it is also the nitrogen donor for several key metabolic enzymes and for the de novo synthesis of both purines and pyrimidines (Fig. 2). Serine also participates in the de novo synthesis of nucleotides by serving one carbon unit. Functional genomics further indicates that serine biosynthesis pathway is significant for breast cancer event, which can be attributable to the overexpression of phosphoglycerate dehydrogenase (PHGDH) that controls the flow of intermediates originated from glycolysis (64). Inhibition of PHGDH in cells can result in lower serine and decrease cellular proliferation in vitro. However, this remains unclear in gastric cancer. Tryptophan and its downstream metabolites (mainly including kynurenine, kynurenic acid, anthranilic acid, nicotinic acid) via kynurenine pathway are related to the pathogenesis and prognosis of various malignancies including gastric cancer (65,66). Kynurenine pathway catalyzed by indoleamine-2, 3-dioxygenase (IDO) plays a key role in adapting the tumor microenvironment to favor cancer progression because higher IDO expression is associated with an increase in immunosuppressive T-regulatory cell activity (67), and its immunosuppressive role inhibits T-cell mediated cytotoxicity and cell proliferation of gastric cell lines in vitro (68). Additionally, 3-hydroxyanthanilie acid (downstream metabolites in kynurenine pathway) also has suppressive effects on inflammation and immune response (69). Glycine used in living organism as building blocks of purines is strongly correlated with the rapid proliferation rates, and then antagonizing glycine uptake and its mitochondrial biosynthesis preferentially impair rapidly proliferating cells (70). The indirect anti-angiogenic impact of glycine is also identified in vitro (71,72) and in vivo (73,74), possibly because it might inhibit the proliferation of vascular endothelial cells, finally leading to angiogenesis (74). Elevated proline in tumor tissues might begin with the activation of MMPs and degradation of micro-environmentally extracellular matrix (ECM), subsequently the degradation of collagen catalyzed by proline dehydrogenase (PRODH) that can be regulated under conditions of nutrient stress linked to mTOR signaling system (75). Other elevated amino acids, such as tyrosine, valine and cysteine, can be converted into the TCA intermediates (except for citric acid, isocitrate, succinyl-CoA, oxaloacetate) to generate energy (Fig. 2).

Lipid metabolism

The notable feature of lipid metabolism in cancer cells is an increased rate of lipogenesis and the upregulation of mitochondrial fatty acid β-oxidation, gastric cancer shows a similar tendency and presents typical changes regarding various metabolites involving in lipid metabolism.

Fatty acids, such as hexadecenoic acid, docosahexaenoic acid, eptanoic acid and β-hydroxybutyrate, are significantly larger in gastric cancer tissues than in benign tissues (like chronic superficial gastritis) (28). Octadecanoic acid is also found to be elevated in blood specimens obtained from gastric cancer patients (39). Of them, β-hydroxybutyrate is the common product of fatty acid degradation via β-oxidation, suggesting more intensive decomposition of fatty acids in microenvironment. The accelerated metabolism from lipids to fatty acids and finally ketone bodies consumes fat, which might explain the fact that patients become very thin in later stages of gastric cancer. This signature also has been identified by the xenograft animal models with gastric cancer showing elevated levels of glycerol and hexadecanoic acid (30), resulting from the high activation of adipocyte lipolysis in cancer cells as well as enhanced expression and function of adipocyte hormone-sensitive lipase in cancer cachexia (76). In contrast, some data show that unsaturated fatty acids such as 9-hexadecenoic acid, cis-vaccenic acid, arachidinic acid, hexadecanoic acid and 3-hydroxybutanoic acid are found to be significantly decreased in cancer tissue samples (26). Of note, the level of O-acetylcarnitine, which increases the β-oxidation of fatty acid, shows a declining trend as the early gastric cancer progresses into advanced stage (31). Accordingly, it seems that decreased O-acetylcarnitine might explain the impaired fatty acids β-oxidation in stage III/IV gastric cancer, which is characterized by the decline in unsaturated fatty acids we discussed above (9-hexadecenoic acid, cis-vaccenic acid, arachidinic acid, hexadecanoic acid and 3-hydroxybutanoic acid). However, this discrepancy between different research needs further elucidation with larger samples and different analytical methods. On the other hand, free fatty acids in plasma, including palmitic acid, stearic acid, 9-(Z)-hexadecenoic acid, oleic acid, linoleic acid, docosahexaenoic acid and arachidonic acid, are equivalent in both gastric cancer and gastric benign disorders (25). Therefore, it infers that free fatty acids in blood might be not utilized by tumor cells.

Upregulated lipid peroxides are also confirmed in this disease. Accumulation of 4-hydroxyphenylacetate resulting from the oxidative degradation of lipids was observed in the study of Jung et al (31). Elevation of azelaic acid in blood samples, which is the end product of linoleic acid when subjected to peroxide decomposition (77) and can serve as a marker of lipid peroxidation (78), as observed by Yu et al (25).

Based on that indicated above, these signatures show that cancer cells utilize massive fatty acids to meet the demand of cell membrane synthesis, mainly for lipid raft and lipid-modified signaling molecules (79); and a large fraction of their membrane lipids are biosynthesized de novo rather than scavenging from extracellular sources. In de novo lipogenesis, fatty acid synthase (FAS) catalyzes the synthesis of palmitate from acetyl-CoA or malonyl-CoA in the presence of NADPH as a redox equivalent. FAS expression is commonly low in non-proliferating cells that typically import lipids from the extracellular milieu. In contrast, actively proliferating cells, especially tumor cells, have increased demands for lipids, which is highly dependent on de novo synthesis. So FAS is frequently upregulated in many types of tumors (8082) including gastric cancer (83,84); and increased FAS expression is linked to tumor proliferation, chemoresistance and poorer prognosis in cancers (8588). Thus, this key enzyme implicated in lipogenesis has been studied as potential target in anti-neoplastic therapy (84). On the other hand, enhancement of fatty acid-β oxidation is also considered to be an important metabolic reprogramming in the early stage of some cancer types (89), as it produces more ATP and acetyl coenzyme A which in turn can accelerate the rate of citric acid oxidation and serve as the energy source (90). Furthermore, production of polyunsaturated fatty acids, to some extent, is also associated with tumor cell proliferation, apoptosis and angiogenesis (91,92).

Nucleotide metabolism

Tumor cells are in a state of such rapid proliferation and differentiation that frequent nucleotide synthesis and metabolism are upregulated significantly. Accumulation of the end products of nucleotide catabolism is characterized by the higher levels of uric acid or urate (25,30) in gastric cancer patients or animal models. Other purines compounds like hypoxanthine and guanosine were also increased (35,37), but Aa et al showed decreases in uridine (an RNA building block) (28). Nucleotides are also associated with energy metabolism, mainly in the form of ATP and GTP. Of tumor cells, adequate energy should be supplied to meet their proliferation. In this way, it is assumed that nucleotide phosphates should increase in cancer tissues compared with normal tissues. However, Hirayama and colleagues (35), identified that there was no noticeable difference between gastric cancer tissues and adjacent normal tissues with regard to most nucleotide phosphates (ATP, ADP, GTP, and GDP), total adenylate and energy charge. Accordingly, it infers that cancer cells gain growth superiority over their normal counterparts by switching metabolic patterns of energy to anaerobic glycolysis and possibly fumarate respiration that we have discussed above, instead of securing more ATP.

Other altered metabolisms

Except for the changed metabolisms mentioned above, other metabolite concentrations also show increased or decreased trend in the development of gastric cancer. Increased level of creatinine, a waste product of muscle metabolism, was detected in urine samples of tumor groups (33), which might be induced by lower total body skeletal mass among cachectic patients (93,94). Changes in inositol level of gastric malignancy patients are investigated in either tumor tissues (28,36,37) or urine samples (30), but its mechanism and significance are poorly understood.

3. Metabolomics in diagnosis, treatment and prognostic prediction of gastric cancer

Diagnosis

Early diagnosis is the key element determining the outcome of treatment in cancer research, but current application of cancer biomarkers, endoscopy and imaging is still not satisfactory. Serum biomarkers, like CEA and CA19-9, are not effective given their poor sensitivity or specificity. Inconsistent diagnostic efficacy at endoscopy that results from the variations in skill and experience of endoscopists and pathologist might lead to missed diagnosis at early phase, while positive results displayed on imaging examination (such as barium meal and computer tomography) are prone to advanced stage. Interestingly, utility of various -omics technologies open a new field to discover potential biomarkers for gastric cancer diagnosis, especially based on metabolomics.

Exploration of gastric cancer biomarkers in blood or urine is more appreciated because of its non-invasive priority. Yu et al demonstrated that metabolic profiles were quite different in gastric cancer patients with different pathological types in the Correa model, but intestinal metaplasia shared similar metabolic phenotype (threonate, glutamate and azelaic acid) in plasma with neoplastic groups (25,95). Ikeda et al also identified that there were obvious variations in serum metabolic profiles of gastrointestinal cancers (including esophageal, gastric and colorectal) in contrast to healthy volunteers (27). In particular, changes in the levels of 3-hydroxypropionic acid and pyruvic acid were sufficient to differentiate gastric cancer from esophageal and colorectal cancer, and showed high values for both sensitivity (84.6 and 70.0%) and specificity (71.4 and 90.0%) compared with conventional biomarkers (CA19-9 and CEA) (27). The diagnostic potential of serum metabolic profiles between gastric cancer and non-cancer groups was also confirmed by Song et al, and these alterations occurred at early stage of gastric carcinogenesis (26).

Recently, one urine metabolomics in gastric cancer found that 14 out of 17 metabolites detected from training set (94 urine samples) via GC-MS showed diagnostic value better than classic blood biomarkers on validation set (199 urine samples) (34). Six of them (L-alanine, L-isoleucine, L-serine, L-threonine, L-proline and L-methionine) revealed satisfactory diagnostic values with the area under the ROC of >0.75. Chan et al also revealed that gastric cancer has a unique urine metabolic profiles in contrast to benign gastric diseases and healthy patients, especially 2-hydroxyisobutyrate, 3-indoxylsulfate and alanine, producing a discriminatory model with the area under the curve (AUC) of 0.95 (33). Another study reported that metabolites altered in urinary data of gastric cancer patients was predicted with higher sensitivity than CA19-9 and CEA (31).

In tissue testing, Wu and colleagues indicated that 18 metabolites were detected differently between the malignant tissues and the adjacent non-malignant tissues of gastric mucosa with AUC value of 0.9629 (37), but tissue testing was not a non-invasive approach in contrast to blood or urine testing. Our data, on the other hand, showed that higher levels of tyrosine, phenylalanine and tryptophan in the gastric juice were detected in the early phase of gastric carcinogenesis (40), and the sensitivity and specificity for gastric cancer detection with phenylalanine was 87.9 and 79.4% respectively (41).

Metastasis and prognosis

Most gastric cancer-related deaths occur as a result of metastasis, even among patients undergoing gastrectomy. Unfortunately, no molecular markers for predicting metastasis and prognosis are accessible.

Based on metabolomics, Wu and colleagues showed that five metabolites (increased L-cysteine, hypoxanthine and L-tyrosine; decreased phenanthrenol and butanoic acid) were detected differently between non-invasive (T1 and T2) and invasive (T3 and T4) groups, furthermore, 4-hydroxyphenyl-acetate, alanine, phenylacetylglycine, mannitol, glycolate and arginine levels were significantly correlated with cancer T stage (37). By establishing animal models with gastric cancer cell line SGC-7901, Chen et al confirmed that metabolites correlated to proline and serine metabolism could distinguish metastatic from non-metastatic specimens with an AUC value of 1.0 (36). Study conducted by Hu et al suggested that decreased levels of alanine, glycerol, L-proline, butanoic acid and L-threonic acid as well as increased levels of butanediotic acid and myo-inositol could detect non-metastatic and metastatic groups (AUC=1.00) (30).

Significantly, Chen and coworkers recently evaluated the prognostic value of 17 urinary metabolites, which have been identified differently between gastric cancer group and normal group, by following up 82 out of 112 gastric cancer cases for 3–5 years after surgery (34). They discovered that patients with higher levels of proline, p-cresol and 4-hydroxybenzoic acid display poor prognosis with median survival time 16, 15 and 15 months, respectively. Furthermore, the concentration of p-cresol closely correlated with gastric cancer stage, which was gradually increased with the stage of the patients.

It is possible that changes in proline might be essential in tumor metastasis. As we have mentioned above, proline in tumor tissues might result from the degradation of collagen (73). This process mainly begins with the activation of MMPs and degradation of microenvironmental ECM, which partially accounts for the tumor invasion and metastasis (96). In this respective, elevated proline serving as metastatic biomarker for gastric cancer is possible, but further research is necessary.

Treatment

Chemosensitivity prediction that aims to maximize the therapeutic response and minimize adverse effects is a difficult task in the treatment of advanced tumors. One of classical approaches for predicting the activity of anticancer agents is cell culture testing, which is mainly based on clone formation, cell metabolic activity assays, proliferation and tumor growth in vitro experiments. However, it must be noted that these methods still fail to fully reproduce the tumor microenvironment, although current patient-derived primary cell culture or patient-derived tumor xenograft models are able to retain cellular heterogeneity of original tumors (97).

Lu et al, in particular, suggested that some conventional cytotoxic anticancer agents (vincristine, taxol, 5-fluorouracil, doxorubicin, cisplatin, camptothecin) lost their efficacy apparently when cultured PNAC-1 cells (pancreatic cancer) in vitro were deprived of glucose (98). Similarly, a recent study also identified that high glucose conditions promoted SGC-7901 proliferation in vitro and reduced chemosensitivity in vivo or in vitro (99). We could speculate that responses of gastric cancer against anticancer drugs in actual microenvironment in vivo might be considerably different from what we expect in culture condition. Therefore, utilizing metabolomics is considered to be a promising tool to assess the sensitivity of chemotherapy in virtual conditions and discovering therapeutic targets regarding specific tumor metabolism (20,21).

Wang et al applied high performance liquid chromatography coupled with a quadrupole time-of-flight mass spectrometer to predict chemotherapy response in a human xenograft model of gastric cancer administered with cisplatin plus 5-fluorouracil (5-FU) (100). Consequently, 1-acyl-lysophosphatidylcholine and polyunsaturated fatty acid were proposed to surveil gastric cancer chemosensitivity, since 1-acyl-lysophosphatidylcholine can regulate the activity of enzymes like phospholipase A2 (PLA2) and lysophosphatidylcholine acetyltransferases. PLA2 catalyzes the production of arachidonic acid that is likely to promote cell cycle arrest and apoptosis dependent on ceramide pathway (101,102), while lysophosphatidylcholine acetyltransferases catalyzes phospholipid synthesis linked to tumor cell proliferation. Another study suggested that proline was reduced while glutamate increased dramatically, and PRODH (catalyzes the metabolic production of glutamate from proline proceeds) mRNA expression was upregulated 2-fold after 5-FU administration; but they were less affected in 5-FU-resistant cells (103). Thus PRODH might make it possible to be a marker for assessing intracellular dynamic responses to 5-FU. Additionally, Kim and colleagues utilized 1H-NMR to investigate the metabolic changes in urine sample following Adriamycin (ADR) treatment for gastric adenocarcinoma in an animal model (104). This study revealed that levels of trimethylamine oxide, hippurate and taurine, which all decreased in tumor group without treatment, were increased dramatically after ADR disposal; while 2-oxoglutarate, 3-indoxylsulfate, trigonelline, trimethylamine and citrate recovered to those of normal group (104). Alterations in these metabolites might be ascribed to the pharmacological activity of ADR that activates apoptotic process of gastric cancer cells via ADR-induced genotoxic stress.

In another study, dysregulation of pyruvic acid efflux in gastric cardia cancer was observed with the combination of proteomics and metabolomics (38). Furthermore, Cai et al also found that downregulation of lactate dehydrogenase A (LDH-A) and overexpression of pyruvate dehydrogenase B (PDH-B) could force pyruvic acid into the Krebs cycle rather than the glycolysis process in gastric cancer cell line AGS, consequently inhibiting cell growth and migration (38). In view of the above, LDH or PDH might serve as a therapeutic target in gastric cancer treatment.

4. Current perspectives and future directions

As we indicated above, cumulative studies employing metabolomics have yielded initial and promising results in gastric cancer research. However, inconsistent results across studies can be observed, probably because of the different sensitivity of metabolomics methods (105), variety of experimental subjects (patients, animal models or in vitro cell culture), and the number of samples. Additionally, values of those biomarkers should be further validated with larger cohorts and normalized metabolomics analysis. Furthermore, it should be noted that investigations targeted at the mechanism of the altered metabolism and specific metabolic pathways in gastric cancer are relatively deficient at present, so it is difficult to draw a clear dividing line on metabolism for common cancers and this disease based on a handful of studies that looked also at the role of metabolomics. Overall, exploring the metabolic disorders and gastric carcinogenesis still has far to go.

On the other hand, metabolomics locate at the downstream of genomics, transcriptomics and proteomics, mapping the complete metabolic changes under specific conditions associated with pathogenic factors, host or environmental co-effectors. However, it is essential to combine metabolomics with other-omics methods to get a more integrated understanding of gastric carcinogenesis (Fig. 1). For instance, metabolomic genome-wide association studies (mGWAS) have their priority in quantifying metabolic data and uncovering genetic variants affecting metabolite levels (106). Impacts of the microbiome on the metabolome are also an area of increasing interest, because perturbation of gastrointestinal microbiota composition or function including Helicobacter pylori has been proved to play a role in gastric carcinogenesis (107,108). Furthermore, microbe-derived metabolites also produce effects on cancer cells, such as butanoic acid. Some research revealed that it can modulate immune response via the differentiation of colonic regulatory T cells (109) and inhibit colonic tumor cells (110,111), although the signaling mechanism was not clearly understood. Thus, it can explain the fact that some changed metabolites in gastric cancer such as butanoic acid (37), mannitol (37) and p-cresol (34) that are commonly thought of artificial substances, can originate from fermentation by microorganism in gastric flora. Given this, it is reasonable to presume that gastric flora might be incorporated into an in-depth study of the prominent disorders of metabolism in gastric cancer, but there is still a gap in further research.

5. Conclusions

Gastric cancer is one of the most malignant tumors worldwide, and remains a major global health threat. Though its pathogenesis is unknown, promising discoveries have been made with the emergence of -omics studies. Most strikingly, metabolomics provides us in-depth information on metabolic perturbation between healthy and neoplastic states in the stomach, and further help us discovery disease-specific biomarkers. As technology advances and our understanding of metabolic perturbation in gastric cancer grows, new diagnostic and therapeutic targets will undoubtedly emerge. Ultimately, these advances can be translated into clinical practice to realize the goal of truly personalized cancer treatment.

Acknowledgments

This study was supported by National Natural Science Foundation of China (no. 81672410).

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2016. CA Cancer J Clin. 66:7–30. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Conteduca V, Sansonno D, Lauletta G, Russi S, Ingravallo G and Dammacco F: H. pylori infection and gastric cancer: State of the art (Review). Int J Oncol. 42:5–18. 2013.

3 

Amieva M and Peek RM Jr: Pathobiology of Helicobacter pylori-induced gastric cancer. Gastroenterology. 150:64–78. 2016. View Article : Google Scholar

4 

Kim J, Yum S, Kang C and Kang SJ: Gene-gene interactions in gastrointestinal cancer susceptibility. Oncotarget. 7:67612–67625. 2016.PubMed/NCBI

5 

Raei N, Behrouz B, Zahri S and Latifi-Navid S: Helicobacter pylori infection and dietary factors act synergistically to promote gastric cancer. Asian Pac J Cancer Prev. 17:917–921. 2016. View Article : Google Scholar : PubMed/NCBI

6 

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

7 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Yun J, Rago C, Cheong I, Pagliarini R, Angenendt P, Rajagopalan H, Schmidt K, Willson JK, Markowitz S, Zhou S, 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

9 

Xu W, Yang H, Liu Y, Yang Y, Wang P, Kim SH, Ito S, Yang C, Wang P, Xiao MT, et al: Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of α-ketoglutarate-dependent dioxygenases. Cancer Cell. 19:17–30. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Dang L, White DW, Gross S, Bennett BD, Bittinger MA, Driggers EM, Fantin VR, Jang HG, Jin S, Keenan MC, et al: Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature. 465:9662010. View Article : Google Scholar : PubMed/NCBI

11 

Fischer K, Hoffmann P, Voelkl S, Meidenbauer N, Ammer J, Edinger M, Gottfried E, Schwarz S, Rothe G, Hoves S, et al: Inhibitory effect of tumor cell-derived lactic acid on human T cells. Blood. 109:3812–3819. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Dietl K, Renner K, Dettmer K, Timischl B, Eberhart K, Dorn C, Hellerbrand C, Kastenberger M, Kunz-Schughart LA, Oefner PJ, et al: Lactic acid and acidification inhibit TNF secretion and glycolysis of human monocytes. J Immunol. 184:1200–1209. 2010. View Article : Google Scholar

13 

Herber DL, Cao W, Nefedova Y, Novitskiy SV, Nagaraj S, Tyurin VA, Corzo A, Cho HI, Celis E, Lennox B, et al: Lipid accumulation and dendritic cell dysfunction in cancer. Nat Med. 16:880–886. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Zhang J, Ahn WS, Gameiro PA, Keibler MA, Zhang Z and Stephanopoulos G: 13C isotope-assisted methods for quantifying glutamine metabolism in cancer cells. Methods Enzymol. 542:369–389. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Beger RD: A review of applications of metabolomics in cancer. Metabolites. 3:552–574. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Putri SP, Yamamoto S, Tsugawa H and Fukusaki E: Current metabolomics: Technological advances. J Biosci Bioeng. 116:9–16. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Duarte IF, Diaz SO and Gil AM: NMR metabolomics of human blood and urine in disease research. J Pharm Biomed Anal. 93:17–26. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Ussher JR, Elmariah S, Gerszten RE and Dyck JR: The emerging role of metabolomics in the diagnosis and prognosis of cardiovascular disease. J Am Coll Cardiol. 68:2850–2870. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Guma M, Tiziani S and Firestein GS: Metabolomics in rheumatic diseases: Desperately seeking biomarkers. Nat Rev Rheumatol. 12:269–281. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Herrmann K, Walch A, Balluff B, Tänzer M, Höfler H, Krause BJ, Schwaiger M, Friess H, Schmid RM and Ebert MP: Proteomic and metabolic prediction of response to therapy in gastrointestinal cancers. Nat Clin Pract Gastroenterol Hepatol. 6:170–183. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Armitage EG and Southam AD: Monitoring cancer prognosis, diagnosis and treatment efficacy using metabolomics and lipidomics. Metabolomics. 12:1462016. View Article : Google Scholar : PubMed/NCBI

22 

Jayavelu ND and Bar NS: Metabolomic studies of human gastric cancer (Review). World J Gastroenterol. 20:8092–8101. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Chan AW, Gill RS, Schiller D and Sawyer MB: Potential role of metabolomics in diagnosis and surveillance of gastric cancer. World J Gastroenterol. 20:12874–12882. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Miyagi Y, Higashiyama M, Gochi A, Akaike M, Ishikawa T, Miura T, Saruki N, Bando E, Kimura H, Imamura F, et al: Plasma free amino acid profiling of five types of cancer patients and its application for early detection. PLoS One. 6:e241432011. View Article : Google Scholar : PubMed/NCBI

25 

Yu L, Aa J, Xu J, Sun M, Qian S, Cheng L, Yang S and Shi R: Metabolomic phenotype of gastric cancer and precancerous stages based on gas chromatography time-of-flight mass spectrometry. J Gastroenterol Hepatol. 26:1290–1297. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Song H, Peng JS, Dong-Sheng Y, Yang ZL, Liu HL, Zeng YK, Shi XP and Lu BY: Serum metabolic profiling of human gastric cancer based on gas chromatography/mass spectrometry. Braz J Med Biol Res. 45:78–85. 2012. View Article : Google Scholar

27 

Ikeda A, Nishiumi S, Shinohara M, Yoshie T, Hatano N, Okuno T, Bamba T, Fukusaki E, Takenawa T, Azuma T, et al: Serum metabolomics as a novel diagnostic approach for gastrointestinal cancer. Biomed Chromatogr. 26:548–558. 2012. View Article : Google Scholar

28 

Aa J, Yu L, Sun M, Liu L, Li M, Cao B, Shi J, Xu J, Cheng L, Zhou J, et al: Metabolic features of the tumor microenvironment of gastric cancer and the link to the systemic macroenvironment. Metabolomics. 8:164–173. 2012. View Article : Google Scholar

29 

Choi JM, Park WS, Song KY, Lee HJ and Jung BH: Development of simultaneous analysis of tryptophan metabolites in serum and gastric juice - an investigation towards establishing a biomarker test for gastric cancer diagnosis. Biomed Chromatogr. 30:1963–1974. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Hu JD, Tang HQ, Zhang Q, Fan J, Hong J, Gu JZ and Chen JL: Prediction of gastric cancer metastasis through urinary metabolomic investigation using GC/MS. World J Gastroenterol. 17:727–734. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Jung J, Jung Y, Bang EJ, Cho SI, Jang YJ, Kwak JM, Ryu DH, Park S and Hwang GS: Noninvasive diagnosis and evaluation of curative surgery for gastric cancer by using NMR-based metabolomic profiling. Ann Surg Oncol. 21(Suppl 4): S736–S742. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Liang Q, Wang C and Li B: Metabolomic analysis using liquid chromatography/mass spectrometry for gastric cancer. Appl Biochem Biotechnol. 176:2170–2184. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Chan AW, Mercier P, Schiller D, Bailey R, Robbins S, Eurich DT, Sawyer MB and Broadhurst D: (1)H-NMR urinary metabolomic profiling for diagnosis of gastric cancer. Br J Cancer. 114:59–62. 2016. View Article : Google Scholar

34 

Chen Y, Zhang J, Guo L, Liu L, Wen J, Xu L, Yan M, Li Z, Zhang X, Nan P, et al: A characteristic biosignature for discrimination of gastric cancer from healthy population by high throughput GC-MS analysis. Oncotarget. 7:87496–87510. 2016.PubMed/NCBI

35 

Hirayama A, Kami K, Sugimoto M, Sugawara M, Toki N, Onozuka H, Kinoshita T, Saito N, Ochiai A, Tomita M, et al: Quantitative metabolome profiling of colon and stomach cancer microenvironment by capillary electrophoresis time-of-flight mass spectrometry. Cancer Res. 69:4918–4925. 2009. View Article : Google Scholar : PubMed/NCBI

36 

Chen JL, Tang HQ, Hu JD, Fan J, Hong J and Gu JZ: Metabolomics of gastric cancer metastasis detected by gas chromatography and mass spectrometry. World J Gastroenterol. 16:5874–5880. 2010. View Article : Google Scholar : PubMed/NCBI

37 

Wu H, Xue R, Tang Z, Deng C, Liu T, Zeng H, Sun Y and Shen X: Metabolomic investigation of gastric cancer tissue using gas chromatography/mass spectrometry. Anal Bioanal Chem. 396:1385–1395. 2010. View Article : Google Scholar

38 

Cai Z, Zhao JS, Li JJ, Peng DN, Wang XY, Chen TL, Qiu YP, Chen PP, Li WJ, Xu LY, et al: A combined proteomics and metabolomics profiling of gastric cardia cancer reveals characteristic dysregulations in glucose metabolism. Mol Cell Proteomics. 9:2617–2628. 2010. View Article : Google Scholar : PubMed/NCBI

39 

Song H, Wang L, Liu HL, Wu XB, Wang HS, Liu ZH, Li Y, Diao DC, Chen HL and Peng JS: Tissue metabolomic fingerprinting reveals metabolic disorders associated with human gastric cancer morbidity. Oncol Rep. 26:431–438. 2011.PubMed/NCBI

40 

Deng K, Lin S, Zhou L, Geng Q, Li Y, Xu M and Na R: Three aromatic amino acids in gastric juice as potential biomarkers for gastric malignancies. Anal Chim Acta. 694:100–107. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Deng K, Lin S, Zhou L and Li Y, Chen M, Wang Y and Li Y: High levels of aromatic amino acids in gastric juice during the early stages of gastric cancer progression. PLoS One. 7:e494342012. View Article : Google Scholar : PubMed/NCBI

42 

Koukourakis MI, Pitiakoudis M, Giatromanolaki A, Tsarouha A, Polychronidis A, Sivridis E and Simopoulos C: Oxygen and glucose consumption in gastrointestinal adenocarcinomas: Correlation with markers of hypoxia, acidity and anaerobic glycolysis. Cancer Sci. 97:1056–1060. 2006. View Article : Google Scholar : PubMed/NCBI

43 

Pedersen PL, Mathupala S, Rempel A, Geschwind JF and Ko YH: Mitochondrial bound type II hexokinase: A key player in the growth and survival of many cancers and an ideal prospect for therapeutic intervention. Biochim Biophys Acta. 1555:14–20. 2002. View Article : Google Scholar : PubMed/NCBI

44 

Gatenby RA and Gillies RJ: Why do cancers have high aerobic glycolysis? Nat Rev Cancer. 4:891–899. 2004. View Article : Google Scholar : PubMed/NCBI

45 

Yuan LW, Yamashita H and Seto Y: Glucose metabolism in gastric cancer: The cutting-edge. World J Gastroenterol. 22:2046–2059. 2016. View Article : Google Scholar : PubMed/NCBI

46 

Israelsen WJ and Vander Heiden MG: Pyruvate kinase: Function, regulation and role in cancer. Semin Cell Dev Biol. 43:43–51. 2015. View Article : Google Scholar : PubMed/NCBI

47 

Wu J, Hu L, Chen M, Cao W, Chen H and He T: Pyruvate kinase M2 overexpression and poor prognosis in solid tumors of digestive system: Evidence from 16 cohort studies. Onco Targets Ther. 9:4277–4288. 2016. View Article : Google Scholar : PubMed/NCBI

48 

Augoff K, Hryniewicz-Jankowska A and Tabola R: Lactate dehydrogenase 5: An old friend and a new hope in the war on cancer. Cancer Lett. 358:1–7. 2015. View Article : Google Scholar

49 

Le A, Cooper CR, Gouw AM, Dinavahi R, Maitra A, Deck LM, Royer RE, Vander Jagt DL, Semenza GL and Dang CV: 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

50 

Dhup S, Dadhich RK, Porporato PE and Sonveaux P: Multiple biological activities of lactic acid in cancer: Influences on tumor growth, angiogenesis and metastasis. Curr Pharm Des. 18:1319–1330. 2012. View Article : Google Scholar : PubMed/NCBI

51 

Lardner A: The effects of extracellular pH on immune function. J Leukoc Biol. 69:522–530. 2001.PubMed/NCBI

52 

Gottfried E, Kunz-Schughart LA, Ebner S, Mueller-Klieser W, Hoves S, Andreesen R, Mackensen A and Kreutz M: Tumor-derived lactic acid modulates dendritic cell activation and antigen expression. Blood. 107:2013–2021. 2006. View Article : Google Scholar

53 

Chen Z, Lu W, Garcia-Prieto C and Huang P: The Warburg effect and its cancer therapeutic implications. J Bioenerg Biomembr. 39:267–274. 2007. View Article : Google Scholar : PubMed/NCBI

54 

Costello LC and Franklin RB: 'Why do tumour cells glycolyse?': From glycolysis through citrate to lipogenesis. Mol Cell Biochem. 280:1–8. 2005. View Article : Google Scholar

55 

Lu Y, Zhang X, Zhang H, Lan J, Huang G, Varin E, Lincet H, Poulain L and Icard P: Citrate induces apoptotic cell death: A promising way to treat gastric carcinoma? Anticancer Res. 31:797–805. 2011.PubMed/NCBI

56 

Weljie AM and Jirik FR: Hypoxia-induced metabolic shifts in cancer cells: Moving beyond the Warburg effect. Int J Biochem Cell Biol. 43:981–989. 2011. View Article : Google Scholar

57 

Ichinoe M, Yanagisawa N, Mikami T, Hana K, Nakada N, Endou H, Okayasu I and Murakumo Y: L-Type amino acid transporter 1 (LAT1) expression in lymph node metastasis of gastric carcinoma: Its correlation with size of metastatic lesion and Ki-67 labeling. Pathol Res Pract. 211:533–538. 2015. View Article : Google Scholar : PubMed/NCBI

58 

Zhang M, Zhu GY, Gao HY, Zhao SP and Xue Y: Expression of tissue levels of matrix metalloproteinases and tissue inhibitors of metalloproteinases in gastric adenocarcinoma. J Surg Oncol. 103:243–247. 2011. View Article : Google Scholar : PubMed/NCBI

59 

Sampieri CL, León-Córdoba K and Remes-Troche JM: Matrix metalloproteinases and their tissue inhibitors in gastric cancer as molecular markers. J Cancer Res Ther. 9:356–363. 2013. View Article : Google Scholar : PubMed/NCBI

60 

Qian HR and Yang Y: Functional role of autophagy in gastric cancer. Oncotarget. 7:17641–17651. 2016.PubMed/NCBI

61 

Hensley CT, Wasti AT and DeBerardinis RJ: Glutamine and cancer: Cell biology, physiology, and clinical opportunities. J Clin Invest. 123:3678–3684. 2013. View Article : Google Scholar : PubMed/NCBI

62 

Nicklin P, Bergman P, Zhang B, Triantafellow E, Wang H, Nyfeler B, Yang H, Hild M, Kung C, Wilson C, et al: Bidirectional transport of amino acids regulates mTOR and autophagy. Cell. 136:521–534. 2009. View Article : Google Scholar : PubMed/NCBI

63 

Liu W, Le A, Hancock C, Lane AN, Dang CV, Fan TW and Phang JM: Reprogramming of proline and glutamine metabolism contributes to the proliferative and metabolic responses regulated by oncogenic transcription factor c-MYC. Proc Natl Acad Sci USA. 109:8983–8988. 2012. View Article : Google Scholar : PubMed/NCBI

64 

Possemato R, Marks KM, Shaul YD, Pacold ME, Kim D, Birsoy K, Sethumadhavan S, Woo HK, Jang HG, Jha AK, et al: Functional genomics reveal that the serine synthesis pathway is essential in breast cancer. Nature. 476:346–350. 2011. View Article : Google Scholar : PubMed/NCBI

65 

Godin-Ethier J, Hanafi LA, Piccirillo CA and Lapointe R: Indoleamine 2,3-dioxygenase expression in human cancers: Clinical and immunologic perspectives. Clin Cancer Res. 17:6985–6991. 2011. View Article : Google Scholar : PubMed/NCBI

66 

Wiggins T, Kumar S, Markar SR, Antonowicz S and Hanna GB: Tyrosine, phenylalanine, and tryptophan in gastroesophageal malignancy: A systematic review. Cancer Epidemiol Biomarkers Prev. 24:32–38. 2015. View Article : Google Scholar

67 

Bauer TM, Jiga LP, Chuang JJ, Randazzo M, Opelz G and Terness P: Studying the immunosuppressive role of indoleamine 2,3-dioxygenase: Tryptophan metabolites suppress rat allogeneic T-cell responses in vitro and in vivo. Transpl Int. 18:95–100. 2005. View Article : Google Scholar

68 

Zhang R, Li H, Yu J, Zhao J, Wang X, Wang G, Yao Z, Wei F, Xue Q and Ren X: Immunoactivative role of indoleamine 2,3-dioxygenase in gastric cancer cells in vitro. Mol Med Rep. 4:169–173. 2011.PubMed/NCBI

69 

McGaha TL, Huang L, Lemos H, Metz R, Mautino M, Prendergast GC and Mellor AL: Amino acid catabolism: A pivotal regulator of innate and adaptive immunity. Immunol Rev. 249:135–157. 2012. View Article : Google Scholar : PubMed/NCBI

70 

Jain M, Nilsson R, Sharma S, Madhusudhan N, Kitami T, Souza AL, Kafri R, Kirschner MW, Clish CB and Mootha VK: Metabolite profiling identifies a key role for glycine in rapid cancer cell proliferation. Science. 336:1040–1044. 2012. View Article : Google Scholar : PubMed/NCBI

71 

Rose ML, Madren J, Bunzendahl H and Thurman RG: Dietary glycine inhibits the growth of B16 melanoma tumors in mice. Carcinogenesis. 20:793–798. 1999. View Article : Google Scholar : PubMed/NCBI

72 

Amin K, Li J, Chao WR, Dewhirst MW and Haroon ZA: Dietary glycine inhibits angiogenesis during wound healing and tumor growth. Cancer Biol Ther. 2:173–178. 2003. View Article : Google Scholar : PubMed/NCBI

73 

Bruns H, Petrulionis M, Schultze D, Al Saeedi M, Lin S, Yamanaka K, Ambrazevičius M, Strupas K and Schemmer P: Glycine inhibits angiogenic signaling in human hepatocellular carcinoma cells. Amino Acids. 46:969–976. 2014. View Article : Google Scholar : PubMed/NCBI

74 

Bruns H, Kazanavicius D, Schultze D, Saeedi MA, Yamanaka K, Strupas K and Schemmer P: Glycine inhibits angiogenesis in colorectal cancer: Role of endothelial cells. Amino Acids. 48:2549–2558. 2016. View Article : Google Scholar : PubMed/NCBI

75 

Phang JM, Donald SP, Pandhare J and Liu Y: The metabolism of proline, a stress substrate, modulates carcinogenic pathways. Amino Acids. 35:681–690. 2008. View Article : Google Scholar : PubMed/NCBI

76 

Agustsson T, Rydén M, Hoffstedt J, van Harmelen V, Dicker A, Laurencikiene J, Isaksson B, Permert J and Arner P: Mechanism of increased lipolysis in cancer cachexia. Cancer Res. 67:5531–5537. 2007. View Article : Google Scholar : PubMed/NCBI

77 

Raghavamenon A, Garelnabi M, Babu S, Aldrich A, Litvinov D and Parthasarathy S: Alpha-tocopherol is ineffective in preventing the decomposition of preformed lipid peroxides and may promote the accumulation of toxic aldehydes: A potential explanation for the failure of antioxidants to affect human atherosclerosis. Antioxid Redox Signal. 11:1237–1248. 2009. View Article : Google Scholar : PubMed/NCBI

78 

Schallreuter KU and Wood JM: Azelaic acid as a competitive inhibitor of thioredoxin reductase in human melanoma cells. Cancer Lett. 36:297–305. 1987. View Article : Google Scholar : PubMed/NCBI

79 

Muñoz-Pinedo C, El Mjiyad N and Ricci JE: Cancer metabolism: Current perspectives and future directions. Cell Death Dis. 3:e2482012. View Article : Google Scholar : PubMed/NCBI

80 

Swinnen JV, Roskams T, Joniau S, Van Poppel H, Oyen R, Baert L, Heyns W and Verhoeven G: Overexpression of fatty acid synthase is an early and common event in the development of prostate cancer. Int J Cancer. 98:19–22. 2002. View Article : Google Scholar : PubMed/NCBI

81 

Flavin R, Peluso S, Nguyen PL and Loda M: Fatty acid synthase as a potential therapeutic target in cancer. Future Oncol. 6:551–562. 2010. View Article : Google Scholar : PubMed/NCBI

82 

Hao Q, Li T, Zhang X, Gao P, Qiao P, Li S and Geng Z: Expression and roles of fatty acid synthase in hepatocellular carcinoma. Oncol Rep. 32:2471–2476. 2014.PubMed/NCBI

83 

Kusakabe T, Nashimoto A, Honma K and Suzuki T: Fatty acid synthase is highly expressed in carcinoma, adenoma and in regenerative epithelium and intestinal metaplasia of the stomach. Histopathology. 40:71–79. 2002. View Article : Google Scholar : PubMed/NCBI

84 

Ito T, Sato K, Maekawa H, Sakurada M, Orita H, Shimada K, Daida H, Wada R, Abe M, Hino O, et al: Elevated levels of serum fatty acid synthase in patients with gastric carcinoma. Oncol Lett. 7:616–620. 2014.PubMed/NCBI

85 

Lin HP, Cheng ZL, He RY, Song L, Tian MX, Zhou LS, Groh BS, Liu WR, Ji MB, Ding C, et al: Destabilization of fatty acid synthase by acetylation inhibits de novo lipogenesis and tumor cell growth. Cancer Res. 76:6924–6936. 2016. View Article : Google Scholar : PubMed/NCBI

86 

Takahiro T, Shinichi K and Toshimitsu S: Expression of fatty acid synthase as a prognostic indicator in soft tissue sarcomas. Clin Cancer Res. 9:2204–2212. 2003.PubMed/NCBI

87 

Menendez JA, Lupu R and Colomer R: Inhibition of tumor-associated fatty acid synthase hyperactivity induces synergistic chemosensitization of HER-2/neu-overexpressing human breast cancer cells to docetaxel (taxotere). Breast Cancer Res Treat. 84:183–195. 2004. View Article : Google Scholar : PubMed/NCBI

88 

Duan J, Sun L, Huang H, Wu Z, Wang L and Liao W: Overexpression of fatty acid synthase predicts a poor prognosis for human gastric cancer. Mol Med Rep. 13:3027–3035. 2016.PubMed/NCBI

89 

Khasawneh J, Schulz MD, Walch A, Rozman J, Hrabe de Angelis M, Klingenspor M, Buck A, Schwaiger M, Saur D, Schmid RM, et al: Inflammation and mitochondrial fatty acid beta-oxidation link obesity to early tumor promotion. Proc Natl Acad Sci USA. 106:3354–3359. 2009. View Article : Google Scholar : PubMed/NCBI

90 

Liu Y: Fatty acid oxidation is a dominant bioenergetic pathway in prostate cancer. Prostate Cancer Prostatic Dis. 9:230–234. 2006. View Article : Google Scholar : PubMed/NCBI

91 

Hyde CA and Missailidis S: Inhibition of arachidonic acid metabolism and its implication on cell proliferation and tumour-angiogenesis. Int Immunopharmacol. 9:701–715. 2009. View Article : Google Scholar : PubMed/NCBI

92 

Lu X, Yu H, Ma Q, Shen S and Das UN: Linoleic acid suppresses colorectal cancer cell growth by inducing oxidant stress and mitochondrial dysfunction. Lipids Health Dis. 9:1062010. View Article : Google Scholar : PubMed/NCBI

93 

Swaminathan R, Major P, Snieder H and Spector T: Serum creatinine and fat-free mass (lean body mass). Clin Chem. 46:1695–1696. 2000.PubMed/NCBI

94 

Eisner R, Stretch C, Eastman T, Xia J, Hau D, Damaraju S, Greiner R, Wishart D and Baracos V: Learning to predict cancer-associated skeletal muscle wasting from 1H-NMR profiles of urinary metabolites. Metabolomics. 7:25–34. 2011. View Article : Google Scholar

95 

Correa P: Human gastric carcinogenesis: A multistep and multifactorial process - First American Cancer Society Award Lecture on Cancer Epidemiology and Prevention. Cancer Res. 52:6735–6740. 1992.PubMed/NCBI

96 

Brown GT and Murray GI: Current mechanistic insights into the roles of matrix metalloproteinases in tumour invasion and metastasis. J Pathol. 237:273–281. 2015. View Article : Google Scholar : PubMed/NCBI

97 

Lau V, Wong AL, Ng C, Mok Y, Lakshmanan M and Yan B: Drug sensitivity testing platforms for gastric cancer diagnostics. J Clin Pathol. 69:93–96. 2016. View Article : Google Scholar

98 

Lu J, Kunimoto S, Yamazaki Y, Kaminishi M and Esumi H: Kigamicin D, a novel anticancer agent based on a new anti-austerity strategy targeting cancer cells' tolerance to nutrient starvation. Cancer Sci. 95:547–552. 2004. View Article : Google Scholar

99 

Zhao W, Chen R, Zhao M, Li L, Fan L and Che XM: High glucose promotes gastric cancer chemoresistance in vivo and in vitro. Mol Med Rep. 12:843–850. 2015.PubMed/NCBI

100 

Wang X, Yan SK, Dai WX, Liu XR, Zhang WD and Wang JJ: A metabonomic approach to chemosensitivity prediction of cisplatin plus 5-fluorouracil in a human xenograft model of gastric cancer. Int J Cancer. 127:2841–2850. 2010. View Article : Google Scholar

101 

Ilsley JN, Nakanishi M, Flynn C, Belinsky GS, De Guise S, Adib JN, Dobrowsky RT, Bonventre JV and Rosenberg DW: Cytoplasmic phospholipase A2 deletion enhances colon tumorigenesis. Cancer Res. 65:2636–2643. 2005. View Article : Google Scholar : PubMed/NCBI

102 

Ganesan K, Ivanova T, Wu Y, Rajasegaran V, Wu J, Lee MH, Yu K, Rha SY, Chung HC, Ylstra B, et al: Inhibition of gastric cancer invasion and metastasis by PLA2G2A, a novel beta-catenin/TCF target gene. Cancer Res. 68:4277–4286. 2008. View Article : Google Scholar : PubMed/NCBI

103 

Sasada S, Miyata Y, Tsutani Y, Tsuyama N, Masujima T, Hihara J and Okada M: Metabolomic analysis of dynamic response and drug resistance of gastric cancer cells to 5-fluorouracil. Oncol Rep. 29:925–931. 2013.

104 

Kim KB, Yang JY, Kwack SJ, Kim HS, Ryu DH, Kim YJ, Bae JY, Lim DS, Choi SM, Kwon MJ, et al: Potential metabolomic biomarkers for evaluation of adriamycin efficacy using a urinary 1H-NMR spectroscopy. J Appl Toxicol. 33:1251–1259. 2013.

105 

Büscher JM, Czernik D, Ewald JC, Sauer U and Zamboni N: Cross-platform comparison of methods for quantitative metabolomics of primary metabolism. Anal Chem. 81:2135–2143. 2009. View Article : Google Scholar : PubMed/NCBI

106 

Adamski J and Suhre K: Metabolomics platforms for genome wide association studies - linking the genome to the metabolome. Curr Opin Biotechnol. 24:39–47. 2013. View Article : Google Scholar

107 

Lofgren JL, Whary MT, Ge Z, Muthupalani S, Taylor NS, Mobley M, Potter A, Varro A, Eibach D, Suerbaum S, et al: Lack of commensal flora in Helicobacter pylori-infected INS-GAS mice reduces gastritis and delays intraepithelial neoplasia. Gastroenterology. 140:210–220. 2011. View Article : Google Scholar

108 

Lertpiriyapong K, Whary MT, Muthupalani S, Lofgren JL, Gamazon ER, Feng Y, Ge Z, Wang TC and Fox JG: Gastric colonisation with a restricted commensal microbiota replicates the promotion of neoplastic lesions by diverse intestinal microbiota in the Helicobacter pylori INS-GAS mouse model of gastric carcinogenesis. Gut. 63:54–63. 2014. View Article : Google Scholar :

109 

Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, Nakanishi Y, Uetake C, Kato K, Kato T, et al: Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature. 504:446–450. 2013. View Article : Google Scholar : PubMed/NCBI

110 

Vanhoutvin SA, Troost FJ, Hamer HM, Lindsey PJ, Koek GH, Jonkers DM, Kodde A, Venema K and Brummer RJ: Butyrate-induced transcriptional changes in human colonic mucosa. PLoS One. 4:e67592009. View Article : Google Scholar :

111 

Singh N, Gurav A, Sivaprakasam S, Brady E, Padia R, Shi H, Thangaraju M, Prasad PD, Manicassamy S, Munn DH, et al: Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity. 40:128–139. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2017
Volume 51 Issue 1

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
Xiao S and Xiao S: Gastric cancer: Metabolic and metabolomics perspectives (Review). Int J Oncol 51: 5-17, 2017
APA
Xiao, S., & Xiao, S. (2017). Gastric cancer: Metabolic and metabolomics perspectives (Review). International Journal of Oncology, 51, 5-17. https://doi.org/10.3892/ijo.2017.4000
MLA
Xiao, S., Zhou, L."Gastric cancer: Metabolic and metabolomics perspectives (Review)". International Journal of Oncology 51.1 (2017): 5-17.
Chicago
Xiao, S., Zhou, L."Gastric cancer: Metabolic and metabolomics perspectives (Review)". International Journal of Oncology 51, no. 1 (2017): 5-17. https://doi.org/10.3892/ijo.2017.4000