Send Orders for Reprints to [email protected]
804 Current Pharmaceutical Design, 2016, 22, 804-811
A Proteomics Based Approach for the Identification of Gastric Cancer Related Markers
Ishfaq A. Sheikh
1, Zeenat Mirza
1, Ashraf Ali
1, Gjumrakch Aliev
2,3,4and Ghulam Md Ashraf
1,*
1
King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia;
2
GALLY International Biomedical Research Consulting LLC., 7733 Louis Pasteur Drive, #330, San Antonio, TX, 78229, USA;
3School of Health Science and Healthcare Administration, University of Atlanta, E. Johns Crossing,
#175, Johns Creek, GA, 30097, USA;
4Institute of Physiologically Active Compounds Russian Academy of Sciences, Chernogolovka, 142432, Russia
Abstract: Gastric cancer (GC) is the amongst the most common cancer types causing second largest number of cancer related deaths globally. GC is characterized as an aggressive malignancy which is very tough to be detected at an early stage. GC has been defined as a complex, multistep process involving multiple genetic and epigenetic al- terations leading to aberrant expression of key regulating factors. GC according to WHO has been defined as ma- lignant epithelial tumors of the gastric mucosa with glandular differentiation. About one half of the GCs are located
in the lower stomach, and remaining is located in the corpus and fundus of the stomach (20%), lesser curvature (20%), cardia (10%) and greater curvature (3%). GC has been classified into intestinal and diffuse types based on epidemiological and clinico- and histo- pathological features. The etiology of GC is multifactorial and includes dietary as well as non-dietary factors. Despite a lot of research ef- forts, GC remains to be the cancer without clear symptoms at onset, poor prognosis, with metastasis and recurrence. Thus, there is an ur- gent need for identifying novel and diagnostic GC biomarkers and techniques with high sensitivity and specificity. In the present review, we provide a synopsis of proteomics based GC biomarkers discovered from various cancerous specimens such as blood, gastric fluid, tis- sues, cells and H. pylori infected cancer cell lines. The advent of proteomics based GC biomarkers will be a great asset for the early de- tection and treatment of GC.
Keywords: Gastric cancer, proteomics, blood, H. pylori, diagnosis.
INTRODUCTION
Gastric cancer (GC), also called stomach cancer, is one of the deadliest and most common cancers across the world. Currently, it is ranked second in terms of global cancer-related mortality [1, 2].
Due to advancement in various genomic and proteomic techniques, the incidence and mortality have reduced dramatically over the last 50 years in many countries. GC is the fourth most in terms of inci- dence, which varies among various ethnic population and regions of the world [3]. GC is considered as aggressive malignancy, which is difficult to detect at an early stage [4]. A recent assessment based on a national survey reported GC as the second most common fatal cancer in India with a mortality rate of 12.6% [5]. In India, its pres- ence varies across different zones, with GC being is more prevalent in the southern and north-eastern states with Mizoram recording an age-adjusted rate of 50.6 and 23.3 for men and women, respectively [6, 7]. In middle east, GC occurrence varies considerably in differ- ent countries. Its occurrence is very high in Iran (around 26.1/10
5), low in Israel (12.5/10
5) and very low in Egypt (3.4/10
5) [8]. Among GCC, GC has been reported to be higher in Oman than in all other nations (Bahrain, Kuwait, Qatar, Saudi Arabia, and UAE) [9]. The literature evidences of GC in Saudi Arabia is still in nascent stage with some reports from regions of western [10] and southern Saudi Arabia [11].
The development of GC is a complex, multistep process which involves several parameters. Involvement of various genetic and epigenetic factors, such as activation and alteration of oncogenes, tumor suppressor genes, DNA repair genes, cell cycle regulators, and signaling molecules leads to aberrant expression of key factors regulating cell cycle progression, apoptosis, senescence, and
*
Address correspondence to this author at the King Fahd Medical Research Center, King Abdulaziz University, P. O. Box 80216, Jeddah 21589, Saudi Arabia; Tel: +966593594931; E-mail: [email protected]differentiation [12]. WHO has defined GC as “malignant epithelial tumors of the gastric mucosa with glandular differentiation” [13].
Uncontrolled growth in the lining of stomach can lead to incidence of gastric tumors, which can later invade adjoining normal tissues and can further spread to other parts of the body to cause cancer.[14]. Overall 50% of the GCs are located in the lower part of stomach (pyloris and antrum), 20% are located in the corpus and fundus (body) of the stomach, 20% are in the part of lesser curva- ture, 10% at the cardia and 3% at the greater curvature [15].
Accumulating evidence has confirmed the role of H. pylori infection in the pathogenesis of GC besides other factors. H. pylori causes chronic gastritis, the precursor to all the pathophysiological abnormal characteristic of gastric carcinogenesis [16, 17]. Antican- cer research has proclaimed many novel target molecules and tech- niques [18-21]. However, these precursor lesions only develop in a proportion of infected subjects, and do not necessarily progress into invasive cancers. GC is a cancer without clear symptoms at onset, poor prognosis, and metastasis and recurrence [22, 23]. Early diag- nosis of GC urgently requires novel biomarkers and techniques with high sensitivity and specificity.
PATHOLOGICAL CLASSIFICATION OF GC
Based on epidemiological, clinical and histo-pathological fea- tures GC can be classified into intestinal and diffuse types [24, 25].
The intestinal type of GC display components of glandular, solid or intestinal architecture as well as tubular structures [26]. In contrast to intestinal variant, diffuse variant displays single cells or poorly cohesive cluster of cells that infiltrate the gastric wall, often leading to widespread thickening and rigidity of the gastric wall, known as linitus plastica [27]. Histologically, intestinal-type is well differen- tiated while diffuse-type is poorly differentiated. Intestinal-type occurs more commonly in older patients, while diffuse-type is more prevalent in younger patients [28]. Diffuse-type is not well differen- tiated with infiltrating, non-cohesive cells and occurs mostly in
Ghulam Md Ashraf
1873-4286/16 $58.00+.00 © 2016 Bentham Science Publishers
younger patients [29]. Adenocarcinoma, which accounts for 90% to 95% of all gastric malignancies, is the major histological type of GC [28]. The method used to predict prognosis and direct therapeu- tic decisions of patients with GC across the world is considered as TNM (Tumor Node Metastasis) staging system [30].
ETIOLOGY OF GC
There are several factors, dietary and non-dietary, which con- tribute in the etiology of GC [6]. Factors such as H. pylori infection, increase in obesity, certain dietary factors, such as higher intake of salty foods and smoked meats, consumption of food preserved with nitrites or nitrates, smoking and alcohol abuse is responsible for intestinal-type of GC. It is believed to arise through a long term multistep progression from chronic gastritis to chronic atrophy to intestinal metaplasia to dysplasia [6]. The diffuse-type of GC is more likely to have a genetic cause. The primary genetic etiology, known as hereditary diffuse GC (HDGC), is mainly due to the E- cadherin (CDH1) germline mutation [26, 31]. Studies of GC during last few decades have shown that the etiolology results from the complex interactions between gene and environment [32, 33]. New high-throughput techniques, such as NGS, have revealed its hetero- geneous milieu, such as alterations of many genes, deregulation of signaling pathways, aberrant DNA methylation patterns, and chro- mosomal imbalances [29]. Although there is vast advancement in diagnosis and treatment, the survival rate of GC in five years has been found to be only 20 per cent [6, 34].
GC BIOMARKERS
In order to improve the understanding and behavior of GC and to diagnose it early, a lot of work has been done to search tumor biomarkers. The aim is to improve cure rates by early detection and diagnosis of such tumors. The biomarkers are defined as biological variables that correlate well with clinico-biological outcome. The current cancer biomarker discovery targets are abnormally ex- pressed proteins, and now they are becoming increasingly popular [35]. Carcino-embryonic antigen, CA 19-9, CA 50 and CA 72-4 are presently used biomarkers, but, the sensitivity and specificity of these biomarkers are not sufficient to detect early stage GT. The disadvantages of these biomarkers are that their specificity is not strong and they have low positive rate [36]. To enhance the diag- nostic sensitivity and specificity, few studies have simultaneously evaluated more than one candidate biomarker, leading to the belief that no single biomarker is likely to prove complete prediction [37, 38]. Discovered markers may also define a subset of networked causal proteins that regulate disease phenotype [39]. Regardless of the method and protocol and data's quality, it is possible that some discovered biomarkers may behave as false positives. Therefore, it is must to validate identified biomarkers against definitive biomedi- cal ground to make them more authentic. Validation of biomarker can be executed by number of ways. They can be validated on bench-side in traditional labs, and web based electronic resources such as text-mining, gene ontology, and clinical trials [40].
Advancement in genomics and proteomics technologies have revolutionized the biomarker discovery. Several recent studies re- veal that genomics and proteomics are powerful tools for tumor identification and classification. Identification of genes or proteins expressed in different cancers may serve as predictive, diagnostic, or prognostic biomarkers [41, 42]. Overall, molecular phenotyping of GC is still in its infancy and the search for novel diagnostic, prognostic and predictive biomarkers continues [43]. Identifying single discriminating biomarker (e.g. COX-2, c-myc, p27 or p53) cannot have a major impact, as a single biomarker is likely insuffi- cient for confirmed diagnosis or for making clinical decisions. Dis- covery of miRNA has provided landmark in cancer research.
Analysis of cancer tissues has revealed the importance of miRNAs as molecular markers for cancer classification, prognosis, and drug- response prediction. As a circulating markers, miRNAs can help in
early diagnosis and follow-up investigations in cancer biomarker discovery [44]. Thanks to high stability and the availability of as- says able to quantify their level, miRNAs could be added to the panel of potential biomarkers [45, 46]. Cancer cells act cleverly as they employ multiple and diverse survival pathways, so there is need to define a battery of biomarkers (complex signatures that define multiple outcomes) which can identify those abrupted signal- ing pathways [47]. Such signatures (battery of biomarkers) may more appropriately represent the breadth of molecular diversity inherent in different cancers. They can pave the pathway to under- stand the impact of both nature and nurture on the molecular genet- ics of gastric carcinogenesis. In the coming era of personalized medicine, genomic and proteomic profiling attempts for mining novel biomarkers of GC may provide significant basis for individu- alized treatment to cancer patients.
PROTEOMICS BASED GC BIOMARKERS
According to report produced by WHO in 2008 around 7.6 million people across world die of cancer each year, and it is as- sumed that by 2030, the deaths from cancer will rise to more than 11 million [48, 49]. Based on 2008-2012 data, the number of GC cases increased to 7.4 per 100,000 men and women per year and the number of deaths being 3.4 per 100,000 men and women per year [50]. Consumption of nitrate- or nitrite-rich food (grilled and baked food, salted, or pickled foods) are some of the major well known GC causing factors [51]. Presence of H. pylori infection [52], age (>60 years), a history of earlier stomach disorders [53] are the other causative factors. Proteomics based biomarkers can help in the accurate diagnosis and distinction of GC patients from healthy indi- viduals or patients without GC [54, 55]. Several advanced pro- teomics techniques are used to identify potential target proteins that can act as novel GC biomarkers. Various proteomics techniques such as 2-DE, Gel Electrophoresis, SDS PAGE, iTRAQ, ICAT, Protein Chip array, hydrophilic interaction liquid chromatography (HILIC), LC-MS, GC-MS, HPLC, affinity chromatography and 2- D LC etc were used for identification of cancer biomarkers [56].
The identity of the separated proteins is then confirmed by Mass Spectrometry techniques [57]. Other latest techniques like western blotting and immunohistochemistry (IHC), is used to validate such isolated compounds [58]. Our previous publications have estab- lished galectins as an important proteomics based GC biomarkers with significant changes in their expression during malignancies including GC [59-66]. In the present review, we have tried to ex- plore the recent progress made in biomarker discovery for Gastric Cancer. We have tried to give idea of various proteomics tools used in biomarker discovery. Our aim is to provide a useful guide that predicts the future direction of the advancement of proteomic tech- nology in cancer biomarker discovery.
SPECIMENS USED FOR IDENTIFICATION OF GC BIO- MARKERS
Cancerous Blood Samples
As blood is an easily accessible and easy-to-use clinical speci-
men for most diseases, it is also used for identification of GC bio-
marker. Due to easy accessibility compared to tissue and gastric
fluid, it can be used as useful source for discovering a highly effec-
tive screening biomarker for GC. The prime objective of proteomic
researchers is to identify blood based GC biomarkers using several
advanced proteome technologies. [67]. For initial and advanced
stage analysis of serum ES-MS, MALDI-TOF MS, GC MS and 2-
DE techniques are utilized [68], besides using other sophisticated
techniques like iTRAQ and liquid chromatography techniques (LC-
MS/MS) for plasma analysis at different stages of gastric cancers
[69]. The identified expected biomarkers were compared with se-
rum protein profiles of other types of cancer, to check its specificity
in gastric carcinogenesis. Their cross reactivity with other known
biomarkers were also checked [70, 71]. MKN45 and SC-M1 mouse
xenograft models have been used to check the progression of Gas- tric Carcinogenesis by analyzing their protein profiling [69, 72].
Several proteomics approaches like SELDI-TOF-MS, Protein Chip array HPLC-ESI-MS, hydrophilic interaction liquid chromatogra-
phy (HILIC), and 2-D LC (LC-MS) etc has been combined for thesearch of future biomarkers with high sensitivity and high specific- ity [73-77]. All potential blood based biomarkers identified by dif- ferent proteomics technologies for gastric carcinogenesis can be categorized into future diagnostic biomarker [78]. Some of the probable GC biomarkers obtained by proteomics technology are,
apolipoprotein C-I (apoC-I) and apoC-III [77], thrombin light chainA [79], and three protein masses with 1468, 3935, and 7560m/z [74]. These potential blood based biomarkers have high sensitivity (>89.9%) and high specificity (>71%). Other biomarkers have lesser specificity such as MIF [80] and a 2209-Da peptide [81], can also be considered as future blood biomarkers for GC (>28% sensi- tivity and >83.3% specificity). C9 [69] (sensitivity: 90%; specific- ity: 74%) and IPO-38 [70] (sensitivity: 56.7%; specificity: 93.3%) are additional biomarkers specific for different stages of Gastric Carcinogenesis. Thus, above findings show that blood and serum based biomarkers isolated through different proteomics approaches can be successfully applied to biomarker based drug discovery, using latest diagnostic tools having bigger clinical sample size.
Table 1 shows potential GC biomarkers identified from cancerous blood specimens.
Cancerous Gastric Fluid Specimens
Gastric fluid, which help in food digestion, and provide protec- tion against different infections, can also be used for biomarker discovery particularly in gastric carcinogenesis [82]. Several pro- teomics techniques like Mass Spectrometry (ESI-MS, GC-MS, LC- MS) has been used to separate proteins from gastric fluid [83].
Some interesting results were observed when data from gastric fluid is compared with healthy individual. Patients with gastric ulcers and GC have higher protein concentrations in their gastric juice compared to healthy individuals, but their concentration is lower in patients with duodenal ulcers, than those in healthy individuals [84]. Some of the important GC markers discovered from gastric juice are pepsin-related proteins such as pepsinogen C [83], pepsin
A [83], 1-antitrypsin [84], and pepsinogen II [85], which have been found altered in GC. It can be interpreted that the activation of proteolytic enzymes in gastric juice can have significant implication in GC. Table 2 shows potential GC biomarkers identified from cancerous gastric fluid specimens.
Cancerous Tissue Specimens
Tissue biopsies are major specimen to confirm cases of differ- ent carcinogenesis. Patients with suspected tumors go to surgery, so paired tissues of gastric tumors and surrounding noncancerous gas- tric mucosa obtained after surgical resection are main source of GC confirmation. These samples are usually analyzed by 2-DE com- bined with MALDI-TOF [80, 86], besides other proteomic tech- niques such as ProteinChip arrays [87] and SELDI-TOF-MS [88].
These profiling helps to differentiate tumor from normal tissues or early stages from advanced stages of GC. The primary goal of tis- sue profiling by different proteomics techniques is to discover use- ful therapeutic targets against such diseases. In previous studies, it was observed that expected tissue biomarkers are over expressed in tumor tissues and it is easier to detect individuals with GC from healthy individuals. The biomarkers discovered from tissue speci- men include HER2 (sensitivity: 65%; specificity: 92%) [86], human neutrophil peptides 1–3 (HNPs 1–3), and MIF (P<0.05) [89].
Among them HER2 has been found to be associated with gastric as well as breast cancers [90] and obvious therapeutic target in both cases. The result provides possibility of proteomics technology in identification of such markers which can potentially applied to identification of another cancer. It can check whether known bio- markers in one type of cancer can acts as novel biomarkers for other types of cancers. The expression profiling of proteomics bio- markers like, chloride intracellular channel 1 (CLIC1; low expres- sion correlated with a high 5-year survival rate) [91] in combination with a patient's survival rate and IHC assay results may have a high prognostic value. In brief, the application of different proteomic tools can help in identification and verification of different bio- markers which can provide more robust clinical evaluation of pa- tients with GC. Table 3 shows potential GC biomarkers identified from cancerous tissue specimens.
Table 1. Potential GC biomarkers identified from cancerous blood specimens.
Potential Biomarker Detection Method Verification Method Expression Level References
CF1 2-DE, MALDI-TOF-MS Western blot Down [97]
C9 iTRAQ LC-MS/MS Western blot Up [69]
IPO-38 Antibody microarray ELISA Up [70]
ITIH3 iTRAQ Western blot Up [69]
SLeX HILIC, off-line 2-D LC 2-DE, 2-D DIGE Up [76]
ApoC-I and ApoC-III C8-reverse phase LC-MS/MS ELISA Down [77]
FPA Serum pretreatment with C8 magnetic beads, MALDI-TOF-MS
ELISA Unclear [98]
Thrombin light chain A SELDI-TOF-MS Immunoprecipitation Down [79]
MIF ELISA - Up [80]
a 2209-Da peptide MALDI-TOF-MS Western blot Unclear [81]
Peaks at 1468, 3935, and 7560 m/z
SELDI-TOF-MS, ProteinChip arrays
- Unclear [74]
Cancerous Cells/Cell Line Specimens
Various proteomics and transcriptomics strategy has been ap- plied to discover the molecular details of diseases. Altered protein expression in cell-cell interactions, cell-extracellular matrix adhe- sion, cell motility, proliferation, and tumor immunity, have been explored to solve the puzzle of different cancers [92]. Among them
MET (an oncogene) is found to be involved in the growth and sur- vival of gastric tumor cells [93, 94]. Other proteins such as Vimentin and galectin 1 found in metastatic cells [92] and - enolase (ENO1) in GKN 1-overexpressing cells [95]. These mole- cules can be used as therapeutic targets for GC detection Table 2. Potential GC biomarkers identified from cancerous gastric fluid specimens.
Potential Biomarkers Detection Method Verification Method References
PGC, pepsin A, -defensin ProteinChip arrays, SELDI-TOF-MS ELISA [83]
1-antitrypsin precursor 2-DE MALDI-TOFMS [84]
GKN1, trefoil factor 1, 1-antitryspin, pepsinogen II 2-DE, MALDI LC-MS/MS Western blot [85]
Table 3. Potential GC biomarkers identified from cancerous tissue specimens.
Potential Biomarkers Detection Method Verification Method Expression Level References
Selenium-binding protein 1 2-DE, MALDI-TOF-MS RT-PCR, Western blot, IHC Down [80]
ENO1, GRP78, GRP94, PPIA, PRDX1, PTEN
2-DE, MALDI-TOF-MS Western blot, IHC Down [12]
MAWD-binding protein 2-DE, MALDI-TOF-MS Western blot, Real-Time PCR Down [99]
MAD1L1, HSP27, CYR61 2-DE, MALDI-TOF-MS, YR61 IHC Unclear [100]
CLIC1 2-DE, MALDI-TOF-MS Real-Time PCR, Western blot Up [91]
Cathepsin B 2-DE, MALDI-TOF-MS Western blot, IHC Up [79]
AMP-18 2-DE, MALDI-TOF-MS 2-D Immunoblots Unclear [101]
HSP27 2-DE, MALDI-TOF-MS IHC Up [102]
ATP-dependent RNA helicase DDX39
2-D DIGE, LC-MS/MS Western blot, IHC Unclear [103]
LDHA 2-DE, GC-TOF-MS Real-Time PCR, Western blot Down [88]
PDHB 2-DE, GC-TOF-MS Real-Time PCR, Western blot Up [88]
T3, HIF and fumarate 2-DE, ESI-Q-TOF-MS/MS RT-PCR, Western blot Unclear [104]
CRIP1, HNP-1, S100A6 MALDI imaging MS IHC Unclear [86]
-defensin-1, -defensin-2, S100A8, S100A9
MALDI-TOF-MS HPLC, LC-MS/MS Unclear [88]
PGC ProteinChip arrays, SELDI-TOF-MS IHC Down [87]
HNPs 1–3 and macrophage migra- tion inhibitory factor
ProteinChip arrays, SELDI-TOF-MS LC-MS/MS, IHC Up [89]
Galectin-2 ICAT, LC-MS IHC Unclear [98]
Apo-A1 iTRAQ LC-MS Western blot Down [69]
S100P NanoLC-MS IHC Down [105]
Laminin gamma 2 chain monomer IHC 2-D Immunoblots Up [106]
HER2 IHC, Fluorescence in situ hybridization Imaging mass spectrometry Up [86]
Glycolipid TLC immunostaining GC-MS Down [107]
GM2 TLC immunostaining GC-MS Up [107]
Galectin-2 ICAT, LC-MS Western blot, IHC Down [108]
Table 4. Potential GC biomarkers identified from cancerous cells/cell line specimens.
Cell Line Potential Biomarker Detection Verification Method Expression Level References MkN-1, MkN-
45,KATOIII
GRP78 2-DE, MALDI-TOF/TOF- MS, Western blot, Im-
munoreactions
IHC Unclear [71]
SC-M1, TMC-1 Vimentin, galectin 1 2-D LC-MS/MS, cICAT, Microarray
Western blot, semi- quantitative RT-PCR
Unclear [92]
SNU5 Mitochondrial MET iTRAQ-MS/MS (MET inhibitor treatment)
Western blot Up [109]
GTL16 IL-8, GRO, uPAR, IL-6
Protein arrays (MET in- hibitor treatment)
ELISA Unclear [110]
SGC ENO1 2-DE, MALDI-TOF-MS Western blot, Flow cytometry Down [95]
MKN7, MKN45, HFE145
CTSS iTRAQ-MS/MS IHC, Tissue microarray Up [111]
SNU5, SNU1, AGS, YCC1, KatoIII
phospho-p53 LC-MS/MS Protein antibody array Unclear [109]
SGC7901 15-PGDH 2-DE, Western Blot Unclear [104]
SC-M1 14-3-3 MALDI-TOF-MS IHC Up [112]
SGC and tissues 14-3-3 2-DE Western blot up [113]
RGM-1, AGS Ubiquinol-cytochrome c reductase, mitochondrial
short-chain enoyl- coenzyme A hydratase-
1, HSP60 and EF-Tu
MALDI-TOF-TOF (5-FU treatment) 2-DE, LC-
MS/MS 2-DE
ELISA Western blot
Up [114]
GES-1, SGC7901 Sorcin iTRAQ IHC Up [115]
Table 5. Potential GC biomarkers identified from H. pylori-infected cancerous specimens
1 Potential Biomarker Detection Method Verification Method References
Serum, H. pylori GroES 2-D Immunoblots, Nano LC- MS/MS
Serologic study [96]
Serum, H. pylori 17 antigens 2-D Immunoblots No [116]
Serum, H. pylori 30 antigens 2-D Immunoblots, MALDI-TOF- MS
No [117]
Cells Annexin A2 ProteinChips, SELDI-TOF-MS Western Blot [118]
Cells Serine arginine-rich (SR) proteins (regu- lation of alternative splicing)
SILAC, phosphoprotein enrich- ment, 2-DE, MALDI TOF/TOF-
MS
No [119]
Cells 140 transcripts, 190 protein species 2-DE, MALDI-TOF-MS, cDNA expression array
No [120]
Tissue Annexin A4 2-DE, MALDI-TOF-MS Western blot [121]
H. pylori antioxidant protein group (SodB, KatA, AphC/TsaA, TrxA, Pfr), tricarboxylic
acid cycle proteins (Idh, FrdA, FrdB, FldA, AcnB) and heat shock proteins
(GroEL and ClpB)
2-D DIGE, MALDI-TOF-MS Real-time PCR, DNA mi- croarray
[122]
and treatment. However, for these identified predictive cell bio- markers to be used in clinical settings, rigorous in vivo evaluation of these markers in clinical specimens is necessary. Table 4 shows potential GC biomarkers identified from cancerous cells/cell line specimens.
H. pylori-Infected Cancerous Specimens
Chronic infection with H. pylori is considered a high risk for GC development [52]. For detecting H. pylori-related target pro- teins, H. pylori-infected specimens and H. pylori strains isolated from patients have been used as a source to establish an H. pylori- associated proteomic profile. GroES is considered as H. pylori spe- cific antigen for GC identified by immunoproteomics [96]. Com- parative immunoproteomics can be effective clinical diagnostic tools for H. pylori-infected GC patients. Table 5 shows potential GC biomarkers identified from H. pylori-infected cancerous speci- mens.
CONCLUSION
GC remains to be one of the leading causes of mortality world- wide. Hence due to the high toll of deaths occurring every year, there has always been a compelling need and quest for exploring new ways of diagnosis and treatment. Present work is an attempt to provide an in depth information in an easy and understandable lan- guage about the GC. Although advancements in proteomic research contributed a lot in identifying novel biomarkers for GC diagnosis, but mining miRNAs as biomarkers has been the area of focus in present time. Diagnosing GC using single biomarker does not seem to be sufficient. Although, so far individually large number of bio- markers have been reported, but defining the set of biomarkers could be quite useful in diagnosis and prediction. This review arti- cle gives the full coverage of biomarkers from different sources and assays used for their diagnosis and could be quite useful for clini- cians. This would be helpful in bridging the gap between theoretical knowledge and the experimental assays used in prediction and di- agnosis of GC.
CONFLICTS OF INTEREST
The authors confirm that this article content has no conflict of interest.
ACKNOWLEDGEMENTS
The authors gratefully acknowledge the research facilities pro- vided by King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.
REFERENCES
[1] Pourhoseingholi MA, Vahedi M, Baghestani AR. Burden of gastro- intestinal cancer in Asia; an overview. Gastroenter Hepatol Bed Bench 2015; 8(1): 19-27.
[2] Jing J-J, Liu H-Y, Hao J-K, et al. Gastric cancer incidence and mortality in Zhuanghe, China, between 2005 and 2010. World J Gastroenterol 2012; 18(11): 1262-69.
[3] Rahman R, Asombang AW, Ibdah JA. Characteristics of gastric cancer in Asia. World J Gastroenterol 2014; 20(16): 4483-90.
[4] Qiu F-M, Yu J-K, Chen Y-D, Jin Q-F, Sui M-H, Huang J. Mining novel biomarkers for prognosis of gastric cancer with serum pro- teomics. J Exp Cli Cancer Res 2009; 28(126): 1-7.
[5] Dikshit RP, Mathur G, Mhatre S, Yeole BB. Epidemiological re- view of gastric cancer in India. Ind J Med Paed Oncol 2011; 32(1):
3-11.
[6] Nagini S. Carcinoma of the stomach: A review of epidemiology, pathogenesis, molecular genetics and chemoprevention. World J Gastroenterol 2012; 4(7): 156-69.
[7] Sharma A, Radhakrishnan V. Gastric cancer in India. Ind J Med Paed Oncol 2011; 32(1): 12-6.
[8] Hussein NR. Helicobacter pylori and gastric cancer in the Middle East: A new enigma? World J Gastroenterol 2010; 16(26): 3226-34.
[9] Al-Mahrouqi H, Parkin L, Sharples K. Incidence of stomach cancer in oman and the other gulf cooperation council countries. Oman Med J 2011; 26(4): 258-62.
[10] Al-Radi AO, Ayyub M, Al-Mashat FM, et al. Primary gastrointes- tinal cancers in the Western Region of Saudi Arabia. Is the pattern changing? Saudi Med J 2000; 21(8): 730-4.
[11] Hamdi J, Morad NA. Gastric cancer in southern Saudi Arabia. Ann Saudi Med 1994; 14(3) 195-7.
[12] Bai Y, Fang N, Gu T, Kang Y, Wu J, Yang D, et al. HOXA11 gene is hypermethylation and aberrant expression in gastric cancer. Can- cer Cell Int 2014; 14(79): 1-8.
[13] Dicken BJ, Bigam DL, Cass C, Mackey JR, Joy AA, Hamilton SM.
Gastric adenocarcinoma. Ann Surg 2005; 241(1): 27-39.
[14] Gastric Cancer: The Basics | Oncolink - Cancer Resources http://www.oncolink.org/types/article.cfm?c=152&id=9463.
[15] Dinis-Ribeiro M, Areia M, de Vries AC, et al. Management of precancerous conditions and lesions in the stomach (MAPS): guide- line from the European Society of Gastrointestinal Endoscopy (ESGE), European Helicobacter Study Group (EHSG), European Society of Pathology (ESP), and the Sociedade Portuguesa De En- doscopia Digestiva (SPED). Endoscopy 2012; 44(1): 74-94.
[16] Shi J, Qu Y-P, Hou P. Pathogenetic mechanisms in gastric cancer.
World J Gastroenterol 2014; 20(38): 13804-19.
[17] Watari J, Chen N, Amenta PS, et al. Helicobacter pylori associated chronic gastritis, clinical syndromes, precancerous lesions, and pathogenesis of gastric cancer development. World J Gastroenterol 2014; 20(18): 5461-73.
[18] Tabrez S, Priyadarshini M, Urooj M, et al. Cancer chemoprevention by polyphenols and their potential application as nanomedicine. J Environ Sci Health Part C Environ Carc Ecotox Rev 2013 31(1):
67-98.
[19] Jabir NR, Tabrez S, Ashraf GM, Shakil S, Damanhouri GA, Kamal MA. Nanotechnology-based approaches in anticancer research. Int J Nanomed 2012; 7: 4391-408.
[20] Ali R, Mirza Z, Ashraf GM, et al. New anticancer agents: recent developments in tumor therapy. Anticancer Res 2012; 32(7):2999- 3005.
[21] Alexiou A, Vairaktarakis C, Tsiamis V, Ashraf GM. Application of efficient nanoparticles for early diagnosis and treatment of cancer.
Curr Drug Metabol 2015; 16(8): 662-75.
[22] Saka M, Morita S, Fukagawa T, Katai H. Present and future status of gastric cancer surgery. Jpn J Clin Oncol 2011; 41(3): 307-13.
[23] Wang Z, Yang C, Yang Y, Shen Z, Zhao H, Yao Y. Clinical appli- cation value of bone turnover markers in non-small-cell lung cancer patients with bone metastases. Chin-Ger J Clin Onc 2011; 10(2):
81-4.
[24] Lauren P. The two histological main types of gastric carcinoma:
diffuse and so-called intestinal-type carcinoma. an attempt at a histo-clinical classification. Acta Pathol Microbiol Scand 1965; 64:
31-49.
[25] Hwang JJ, Lee DH, Lee A-R, et al. Characteristics of gastric cancer in peptic ulcer patients with Helicobacter pylori infection. World J Gastroenterol 2015; 21(16): 4954-60.
[26] Lynch HT, Grady W, Suriano G, Huntsman D. Gastric cancer: new genetic developments. J Surg Oncol 2005; 90(3): 114-33.
[27] Bathaie SZ, Miri H, Mohagheghi M-A, Mokhtari- Dizaji M, Shahbazfar A-A, Hasanzadeh H. Saffron aqueous extract inhibits the chemically-induced gastric cancer progression in the wistar al- bino rat. Iran J Basic Med Sci 2013; 16(1): 27-38.
[28] Hu B, El Hajj N, Sittler S, Lammert N, Barnes R, Meloni-Ehrig A.
Gastric cancer: Classification, histology and application of molecu- lar pathology. J Gastrointol Oncol 2012; 3(3): 251-61.
[29] Hudler P. Genetic aspects of gastric cancer instability. Sci World J 2012; 761909: 1-10.
[30] Sousa MTDd, Savassi-Rocha PR, Cabral MMDÁ. Impact of using a lymph node revealing solution in surgical specimens for pathologi- cal staging of gastric cancer. J Brasil Patol Med Lab 2014; 50(6):
445-51.
[31] Kaurah P, Huntsman DG. Hereditary Diffuse Gastric Cancer. In:
Pagon RA, Adam MP, Ardinger HH, et al. Eds. GeneReviews(®).
Seattle (WA): University of Washington, Seattle 1993.
[32] Panani AD. Cytogenetic and molecular aspects of gastric cancer:
clinical implications. Cancer Lett 2008; 266(2): 99-115.
[33] McLean MH, El-Omar EM. Genetics of gastric cancer. Nat Rev Gastroentol Hepatol 2014; 11(11): 664-74.
[34] Karimi P, Islami F, Anandasabapathy S, Freedman ND, Kamangar F. Gastric cancer: descriptive epidemiology, risk factors, screening, and prevention. Cancer Epidemiol Biomarker Prevent 2014; 23(5):
700-13.
[35] Sallam RM. Proteomics in cancer biomarkers discovery: challenges and applications. Dis Markers 2015; 321370: 1-12.
[36] Kobayashi E, Ueda Y, Matsuzaki S, et al. Biomarkers for screen- ing, diagnosis, and monitoring of ovarian cancer. Cancer Epidemiol Biomarker Prevent 2012; 21(11): 1902-12.
[37] Adam BL, Vlahou A, Semmes OJ, Wright GL. Proteomic ap- proaches to biomarker discovery in prostate and bladder cancers.
Proteomics. 2001; 1(10): 1264-70.
[38] Novelli G, Ciccacci C, Borgiani P, Papaluca Amati M, Abadie E.
Genetic tests and genomic biomarkers: regulation, qualification and validation. Clin Cases Min Bone Met 2008; 5(2): 149-54.
[39] Wang Y, Miller DJ, Clarke R. Approaches to working in high- dimensional data spaces: gene expression microarrays. Br J Cancer 2008; 98(6): 1023-8.
[40] Phan JH, Quo C-F, Wang MD. Functional genomics and pro- teomics in the clinical neurosciences: data mining and bioinformat- ics. Prog Brain Res 2006; 158: 83-108.
[41] Mehta S, Shelling A, Muthukaruppan A, Lasham A, Blenkiron C, Laking G, et al. Predictive and prognostic molecular markers for cancer medicine. Ther Adv Med Oncol 2010; 2(2): 125-48.
[42] Tainsky MA. Genomic and proteomic biomarkers for cancer: a multitude of opportunities. Biochem Biophys Acta 2009; 1796(2):
176-93.
[43] Röcken C, Warneke V. Molecular pathology of gastric cancer. Der Pathol 2012; 33 (2): 235-40.
[44] Berger F, Reiser MF. Micro-RNAs as potential new molecular biomarkers in oncology: have they reached relevance for the clini- cal imaging sciences? Theranostics 2013; 3(12): 943-52.
[45] Denk J, Boelmans K, Siegismund C, Lassner D, Arlt S, Jahn H.
MicroRNA profiling of CSF reveals potential biomarkers to detect Alzheimer`s disease. PLoS One 2015; 10(5): 10(5): e0126423.
[46] Lan H, Lu H, Wang X, Jin H. MicroRNAs as potential biomarkers in cancer: opportunities and challenges. BioMed Res Int 2015;
e125094.
[47] Hahn WC, Weinberg RA. Modelling the molecular circuitry of cancer. Nat Rev Cancer 2002; 2(5): 331-41.
[48] Ferlay J, Shin H-R, Bray F, Forman D, Mathers C, Parkin DM.
Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer 2010; 127(12): 2893-917.
[49] Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA: Cancer J Clin 2011; 61(2): 69-90.
[50] Cancer of the Stomach - SEER Stat Fact Sheets.
http://seer.cancer.gov/statfacts/html/stomach.html.
[51] Mirvish SS. Gastric cancer and salivary nitrate and nitrite. Nature 1985; 315(6019): 461-2.
[52] Parsonnet J, Friedman GD, Vandersteen DP, et al. Helicobacter pylori infection and the risk of gastric carcinoma. New Eng J Med 1991; 325(16): 1127-31.
[53] Lo SS, Wu CW, Hsieh MC, Kuo HS, Lui WY, P'Eng FK. Relation- ship between age and clinical characteristics of patients with gastric cancer. J Gastroentol Hepatol 1996; 11(6): 511-4.
[54] Ding X, Yang S, Li W, et al. The potential biomarker panels for identification of major depressive disorder (MDD) patients with and without early life stress (ELS) by metabonomic analysis. PLoS One 2014; 9(5): e97479.
[55] Leal MF, Assumpção PP, Smith MC, Burbano RR. Searching for gastric cancer biomarkers through proteomic approaches. J Gastro- enterol Hepatol Res 2014; 3(3): 989-95.
[56] Cortes DF, Kabulski JL, Lazar AC, Lazar IM. Recent advances in the mass spectrometric analysis of glycoproteins: capillary and mi- crofluidic workflows. Electrophoresis 2011; 32(1): 14-29.
[57] Tuli L, Ressom HW. LC–MS based detection of differential protein expression. J Prot Bioinf 2009; 2: 416-38.
[58] Howat WJ, Lewis A, Jones P, et al. Antibody validation of immu- nohistochemistry for biomarker discovery: Recommendations of a consortium of academic and pharmaceutical based histopathology researchers. Methods 2014; 70(1): 34-8.
[59] Ashraf GM, Banu N, Ahmad A, Singh LP, Kumar R. Purification, characterization, sequencing and biological chemistry of galectin-1 purified from Capra hircus (goat) heart. Prot J 2011; 30(1): 39-51.
[60] Ashraf GM, Bilal N, Suhail N, Hasan S, Banu N. Glycosylation of purified buffalo heart galectin-1 plays crucial role in maintaining its
structural and functional integrity. Biochemistry 2010; 75(12):
1450-7.
[61] Ashraf GM, Greig NH, Khan TA, et al. Protein misfolding and aggregation in Alzheimer's disease and type 2 diabetes mellitus.
CNS Neurol Disord Drug Targets 2014; 13(7): 1280-93.
[62] Ashraf GM, Perveen A, Tabrez S, et al. Altered galectin glycosyla- tion: potential factor for the diagnostics and therapeutics of various cardiovascular and neurological disorders. Adv Exp Med Biol 2015; 822: 67-84.
[63] Hasan SS, Ashraf GM, Banu N. Galectins – Potential targets for cancer therapy. Cancer Lett 2007; 253(1): 25-33.
[64] Ashraf GM, Perveen A, Tabrez S, Zaidi SK, Kamal MA, Banu N.
Studies on the role of goat heart galectin-1 as a tool for detecting post-malignant changes in glycosylation pattern. Saudi J Biol Sci 2015; 22(1): 85-9.
[65] Ashraf GM, Perveen A, Zaidi SK, Tabrez S, Kamal MA, Banu N.
Studies on the role of goat heart galectin-1 as an erythrocyte mem- brane perturbing agent. Saudi J Biol Sci 2015; 22(1): 112-6.
[66] Ashraf GM, Rizvi S, Naqvi S, et al. Purification, characterization, structural analysis and protein chemistry of a buffalo heart galectin- 1. Amino Acids 2010; 39(5): 1321-32.
[67] Lin L-L, Huang H-C, Juan H-F. Discovery of biomarkers for gastric cancer: a proteomics approach. J Prot 2012; 75(11): 3081-97.
[68] Liu W, Liu B, Xin L, et al. Down-regulated expression of comple- ment factor I: a potential suppressive protein for gastric cancer identified by serum proteome analysis. Clin Chim Acta 2007;
377(1-2): 119-26.
[69] Chong PK, Lee H, Zhou J, et al. ITIH3 is a potential biomarker for early detection of gastric cancer. J Prot Res 2010; 9(7): 3671-9.
[70] Hao Y, Yu Y, Wang L, et al. IPO-38 is identified as a novel serum biomarker of gastric cancer based on clinical proteomics technol- ogy. J Prot Res 2008; 7(9): 3668-77.
[71] Tsunemi S, Nakanishi T, Fujita Y, et al. Proteomics-based identifi- cation of a tumor-associated antigen and its corresponding autoanti- body in gastric cancer. Onc Rep 2010; 23(4): 949-56.
[72] Juan H-F, Chen J-H, Hsu W-T, et al. Identification of tumor- associated plasma biomarkers using proteomic techniques: from mouse to human. Proteomics 2004; 4(9): 2766-75.
[73] Ebert MPA, Meuer J, Wiemer JC, et al. Identification of gastric cancer patients by serum protein profiling. J Prot Res 2004; 3(6):
1261-6.
[74] Su Y, Shen J, Qian H, et al. Diagnosis of gastric cancer using deci- sion tree classification of mass spectral data. Cancer Sci 2007;
98(1): 37-43.
[75] Miyagi Y, Higashiyama M, Gochi A, et al. Plasma Free Amino Acid Profiling of Five Types of Cancer Patients and Its Application for Early Detection. PLoS One 2011; 6(9): e24143.
[76] Bones J, Byrne JC, O'Donoghue N, et al. Glycomic and glycopro- teomic analysis of serum from patients with stomach cancer reveals potential markers arising from host defense response mechanisms. J Prot Res 2011; 10(3): 1246-65.
[77] Cohen M, Yossef R, Erez T, et al. Serum apolipoproteins C-I and C-III are reduced in stomach cancer patients: results from MALDI- based peptidome and immuno-based clinical assays. PloS One 2011; 6(1): e14540.
[78] Koevar N, Odreman F, Vindigni A, Grazio SF, Komel R. Proteo- mic analysis of gastric cancer and immunoblot validation of poten- tial biomarkers. World J Gastroenterol 2012; 18(11): 1216-28.
[79] Ebert MPA, Lamer S, Meuer J, et al. Identification of the thrombin light chain a as the single best mass for differentiation of gastric cancer patients from individuals with dyspepsia by proteome analy- sis. J Prot Res 2005; 4(2): 586-90.
[80] Xia HH-X, Yang Y, Chu K-M, et al. Serum macrophage migration- inhibitory factor as a diagnostic and prognostic biomarker for gas- tric cancer. Cancer 2009; 115(23): 5441-9.
[81] Umemura H, Togawa A, Sogawa K, et al. Identification of a high molecular weight kininogen fragment as a marker for early gastric cancer by serum proteome analysis. J Gastroenterol 2011; 46(5):
577-85.
[82] Cooke CL, Torres J, Solnick JV. Biomarkers of Helicobacter py- lori-associated gastric cancer. Gut Microbes 2013; 4(6): 532-40.
[83] Kon OL, Yip T-T, Ho MF, et al. The distinctive gastric fluid pro- teome in gastric cancer reveals a multi-biomarker diagnostic pro- file. BMC Med Genomics 2008; 1(54): 1-14.
[84] Hsu P-I, Chen C-H, Hsieh C-S, et al. Alpha1-antitrypsin precursor in gastric juice is a novel biomarker for gastric cancer and ulcer.
Clin Cancer Res 2007; 13(3): 876-83.
[85] Kam SY, Hennessy T, Chua SC, et al. Characterization of the hu- man gastric fluid proteome reveals distinct pH-dependent protein profiles: implications for biomarker studies. J Proteome Res 2011;
10(10): 4535-46.
[86] Balluff B, Rauser S, Meding S, et al. MALDI imaging identifies prognostic seven-protein signature of novel tissue markers in intes- tinal-type gastric cancer. Am J Pathol 2011; 179(6): 2720-9.
[87] Melle C, Ernst G, Schimmel B, et al. Characterization of pepsino- gen C as a potential biomarker for gastric cancer using a histo- proteomic approach. J Proteome Res 2005; 4(5): 1799-804.
[88] Kim HK, Reyzer ML, Choi IJ, et al. Gastric cancer-specific protein profile identified using endoscopic biopsy samples via MALDI mass spectrometry. J Proteome Res 2010; 9(8): 4123-30.
[89] Mohri Y, Mohri T, Wei W, et al. Identification of macrophage migration inhibitory factor and human neutrophil peptides 1-3 as potential biomarkers for gastric cancer. Br J Cancer 2009; 101(2):
295-302.
[90] Iqbal N, Iqbal N. Human epidermal growth factor receptor 2 (HER2) in cancers: overexpression and therapeutic implications.
Mol Biol Int 2014; e852748.
[91] Chen CD, Wang CS, Huang YH, et al. Overexpression of CLIC1 in human gastric carcinoma and its clinicopathological significance.
Proteomics 2007; 7(1): 155-67.
[92] Chen YR, Juan HF, Huang HC, et al. Quantitative proteomic and genomic profiling reveals metastasis-related protein expression pat- terns in gastric cancer cells. J Proteome Res 2006; 5(10): 2727-42.
[93] Fushida S, Yonemura Y, Urano T, et al. Expression of hepatocyte growth factor(hgf) and C-met gene in human gastric-cancer cell- lines. Int J Oncol 1993; 3(6): 1067-70.
[94] Yonemura Y, Kaji M, Hirono Y, Fushida S, Tsugawa K, Fujimura T, et al. Correlation between overexpression of c-met gene and the progression of gastric cancer. Int J Oncol 1996; 8(3): 555-60.
[95] Yan GR, Xu SH, Tan ZL, Yin XF, He QY. Proteomics characteri- zation of gastrokine 1-induced growth inhibition of gastric cancer cells. Proteomics 2011; 11(18): 3657-64.
[96] Lin YF, Wu MS, Chang CC, et al. Comparative immunoproteomics of identification and characterization of virulence factors from Heli- cobacter pylori related to gastric cancer. Mol cell prot 2006; 5(8):
1484-96.
[97] Liu J, Johnson TV, Lin J, et al. T cell independent mechanism for copolymer-1-induced neuroprotection. Eur J Immunol 2007;
37(11): 3143-54.
[98] Ebert MPA, Röcken C. Molecular screening of gastric cancer by proteome analysis. Eur J Gastroenterol Hepatol 2006; 18(8): 847- [99] 53. Zhang J, Kang B, Tan X, et al. Comparative analysis of the protein
profiles from primary gastric tumors and their adjacent regions:
MAWBP could be a new protein candidate involved in gastric cancer. J Proteome Res 2007; 6(11): 4423-32.
[100] Nishigaki R, Osaki M, Hiratsuka M, et al. Proteomic identification of differentially-expressed genes in human gastric carcinomas.
Proteomics 2005; 5(12): 3205-13.
[101] He Q-Y, Cheung YH, Leung SY, Yuen ST, Chu K-M, Chiu J-F.
Diverse proteomic alterations in gastric adenocarcinoma.
Proteomics 2004; 4(10): 3276-87.
[102] Chen J, Kähne T, Röcken C, et al. Proteome analysis of gastric cancer metastasis by two-dimensional gel electrophoresis and matrix assisted laser desorption/ionization-mass spectrometry for identification of metastasis-related proteins. J Proteome Res 2004;
3(5): 1009-16.
[103] Kikuta K, Kubota D, Saito T, et al. Clinical proteomics identified ATP-dependent RNA helicase DDX39 as a novel biomarker to predict poor prognosis of patients with gastrointestinal stromal tumor. J Proteomics 2012; 75(4): 1089-98.
[104] Liu R, Li Z, Bai S, et al. Mechanism of cancer cell adaptation to metabolic stress: proteomics identification of a novel thyroid
hormone-mediated gastric carcinogenic signaling pathway. Mol Cell Proteomics 2009; 8(1): 70-85.
[105] Jia S-Q, Niu Z-J, Zhang L-H, et al. Identification of prognosis- related proteins in advanced gastric cancer by mass spectrometry- based comparative proteomics. J Can Res Clin Oncol 2009; 135(3):
403-11.
[106] Koshikawa N, Moriyama K, Takamura H, et al. Overexpression of laminin gamma2 chain monomer in invading gastric carcinoma cells. Can Res 1999; 59(21): 5596-601.
[107] Dohi T, Ohta S, Hanai N, Yamaguchi K, Oshima M.
Sialylpentaosylceramide detected with anti-GM2 monoclonal antibody. Structural characterization and complementary expression with GM2 in gastric cancer and normal gastric mucosa.
J Biol Chem 1990; 265(14): 7880-5.
[108] Jung J-H, Kim H-J, Yeom J, et al. Lowered expression of galectin- 2 is associated with lymph node metastasis in gastric cancer. J Gastroenterol 2012; 47(1): 37-48.
[109] Guo T, Zhu Y, Gan CS, et al. Quantitative proteomics discloses MET expression in mitochondria as a direct target of MET kinase inhibitor in cancer cells. Mol Cell Proteomics 2010; 9(12): 2629- 41.
[110] Torti D, Sassi F, Galimi F, et al. A preclinical algorithm of soluble surrogate biomarkers that correlate with therapeutic inhibition of the MET oncogene in gastric tumors. Int J Cancer 2012; 130(6):
1357-66.
[111] Yang Y, Yixuan Y, Lim S-K, et al. Cathepsin S mediates gastric cancer cell migration and invasion via a putative network of metastasis-associated proteins. J Proteome Res 2010; 9(9): 4767- 78.
[112] Tseng C-W, Yang J-C, Chen C-N, et al. Identification of 14-3-3 in human gastric cancer cells and its potency as a diagnostic and prognostic biomarker. Proteomics 2011; 11(12): 2423-39.
[113] Kuramitsu Y, Baron B, Yoshino S, et al. Proteomic differential display analysis shows up-regulation of 14-3-3 sigma protein in human scirrhous-type gastric carcinoma cells. Anticancer Res 2010; 30(11): 4459-65.
[114] Kim HK, Park WS, Kang SH, et al. Mitochondrial alterations in human gastric carcinoma cell line. Am J Phys Cell Phys 2007;
293(2): C761-71.
[115] Deng L, Su T, Leng A, et al. Upregulation of soluble resistance- related calcium-binding protein (sorcin) in gastric cancer. Med Onc 2010; 27(4): 1102-8.
[116] Lahner E, Bernardini G, Possenti S, et al. Immunoproteomics of Helicobacter pylori infection in patients with atrophic body gastritis, a predisposing condition for gastric cancer. Int J Med Microbiol 301(2): 125-32.
[117] Mini R, Bernardini G, Salzano AM, et al. Comparative proteomics and immunoproteomics of Helicobacter pylori related to different gastric pathologies. J Chromatogr B Analyt Technol Biomed Life Sci 2006; 833(1): 63-79.
[118] Das S, Sierra JC, Soman KV, et al. Differential Protein Expression Profiles of Gastric Epithelial Cells Following Helicobacter pylori Infection Using ProteinChips. J Proteome Res 2005; 4(3):920-30.
[119] Holland C, Schmid M, Zimny-Arndt U, et al. Quantitative phosphoproteomics reveals link between Helicobacter pylori infection and RNA splicing modulation in host cells. Proteomics 2011; 11(14): 2798-811.
[120] Backert S, Gressmann H, Kwok T, et al. Gene expression and protein profiling of AGS gastric epithelial cells upon infection with Helicobacter pylori. Proteomics 2005; 5(15): 3902-18.
[121] Lin L-L, Chen C-N, Lin W-C, et al. Annexin A4: A novel molecular marker for gastric cancer with Helicobacter pylori infection using proteomics approach. Proteomics Clin Appl 2008;
2(4): 619-34.
[122] Momynaliev KT, Kashin SV, Chelysheva VV, et al. Functional divergence of Helicobacter pylori related to early gastric cancer. J Proteome Res 2010; 9(1): 254-67.