Skip to main content
Gastroenterology and Hepatology From Bed to Bench logoLink to Gastroenterology and Hepatology From Bed to Bench
. 2015 Winter;8(1):6–18.

Introducing biomarker panel in esophageal, gastric, and colon cancers; a proteomic approach

Mona Zamanian–Azodi 1, Mostafa Rezaei–Tavirani 1, Hadi Hasanzadeh 2, Sara Rahmati Rad 3, Sona Dalilan 1
PMCID: PMC4285927  PMID: 25584171

Abstract

Cancer research is an attractive field in molecular biology and medicine. By applying large-scale tools such as advanced genomics and proteomics, cancer diagnosis and treatment have been improved greatly. Cancers of esophagus, gastric, and colon accounted for major health problem globally. Biomarker panel could bring out the accuracy for cancer evaluation tests as it can suggest a group of candidate molecules specified to particular malignancy in a way that distinguishing malignant tumors from benign, differentiating from other diseases, and identifying each stages with high specificity and sensitivity. In this review, a systematic search of unique protein markers reported by several proteomic literatures are classified in their specific cancer type group as novel panels for feasible accurate malignancy diagnosis and treatment. About thousands of introduced proteins were studied; however, a small number of them belonged to a specific kind of malignancy. In conclusion, despite the fact that combinatorial biomarkers appear to be hopeful, more evaluation of them is crucial to achieve the suitable biomarker panel for clinical application. This effort needs more investigations and researches for finding a specific and sensitive panel.

Key Words: Biomarker panel, Esophagus cancer, Gastric cancer, Colon cancer, Proteomics

Introduction

Cancer is the third foremost cause of death worldwide (1,2). There is no proper early detection and treatment methods available relative to various cancers (3). Gastrointestinal cancers are among the most rampant malignancies and are fatal if remained uncured (4). Common treatments are chemotherapy, radiotherapy, and surgery (5, 6). In 1847, the study of cancer biomarker proteins started by Henry Bence-Jones (7). Mutated proteins as the ultimate products of gene expression, not only related to tumors but also to tumorigenesis (8). These elements can play an integral role in many approaches in cancer studies such as diagnosis, detection, patient monitoring, treatment, and tumor classification (-); pricey, invasive cancer diagnostic tests can be substituted by applying reasonable explicit biomarker test (13). Biomarker test started to demonstrate its novel feature in the detection and management of patients with different malignancies (14,15). High-throughput techniques can be valuable in this field (16). Proteomics as a large-scale protein analyzers is one of the promising tools, enabling a researcher to evaluate malignancies at the molecular stage (17). This method is a new developing ultrasensitive detector while other protein assay tools can just detect ranges about the sub pg mL−1 level (18). Furthermore, in the field of cancer proteomics study, comprehensive analysis of protein interactions, expression, and functions for identifying biomarkers (19), pathways, primary tumors and their metastases relationship and tumor heterogeneity, disease classification, understanding of disease development, and assessment of treatment responses are the main concerns (20, 21).

Sensitivity and specificity in determining biomarkers are crucial due to clinical cancer detection, surveillance after treatment, and therapy selection (22). Protein marker panel could be useful in distinguishing malignant from benign, differentiate from other diseases, and identifying each stages with high specificity and sensitivity. In other words, this preliminary data, if validated in larger clinical studies, could be developed into a serum protein test with less falsely positive diagnoses in which decreases the number of unnecessary biopsies and identifies individuals who need treatment before symptoms begin (23, 24). Since early detection in gastrointestinal cancers is crucial for therapeutic management (25), in this review, selective cancer protein markers and their properties (-) in different common gastrointestinal duct cancers including esophagus, gastric, and colon neoplasm (29, 30) are chosen and discussed for achieving to a proper protein panel for clinical purposes.

Esophageal Cancer

Esophageal cancer as one of the most common malignancies in the world has a wide area broaden from the southern border of the Caspian Sea in Iran across central Asia to China (31). It is accounted for the sixth cause of death in the world with the more than 50 percent metastatic condition at the time of detection (32). The most common type is esophageal squamous cell carcinoma (ESCC) with increasing rate in Western countries (33), due to increase rate of obesity and gastroesophageal reflux disease (GERD) (34). Another type is adenocarcinoma (EAC), which is lethal and occur either within Barrett’s epithelium or in the gastric cardiac mucosa of the distal esophagus (35). Regularly, symptoms start with dysphagia, weight loss, chest pain, pressure or burning, and coughing (-). Many risk factors are accompanied with this malignancy such as diet, smoking, Gastroesophageal reflux disease (GERD), and human papilloma virus (HPV) (-). Methods for detection of esophageal cancer are Esophagoscopy, endoscopic optical, positron emission tomographyraman spectroscopy, immunohistochemistry, and biopsy. The last method (biopsy) is a histological examination for confirming physical and imaging tests (-). General treatment options are photodynamic therapy, chemotherapy, radiotherapy, chemoradiotherapy, and surgery (Esophagectomy), which is the most efficient choice (46). A number of protein markers have been identified for esophageal detection are listed as below (table1).

Table1.

Biomarker panel related to esophagus cancer, the MeSH terms (NBCI Databases) of these markers are mentioned.

Disease Protein name Category Function Condition Application
Esophageal squamous cell carcinoma and
Adenocarcinomas (47)
MAP3K3 protein (48), also known as:
  • MEKK3 protein, human

  • mitogen-activated protein kinase kinase kinase 3, human

  • MAPKKK3 protein, human

Mitogen-activated protein (49) Regulators of nuclear factor kappa B (NF-κB) (48) Up-regulated (47) Early detection (48)
Esophageal squamous cell carcinoma (50)   (Jarid1b) (51), also known as:
  • Jumonji, AT rich interactive domain 1B protein, human

  • RBP2-like protein, human

  • PLU-1 protein, human

  • PLU1 protein, human

  • RBBP2H1A protein, human

  • JARID1B protein, human

  • lysine (K)-specific demethylase 5B, human

Jumonji/Arid1b family (52) Promotes human esophagealcancer cell growth (53) Up-regulated(54) Diagnosis(55)
Esophageal squamous cell carcinoma              (56) UNC-51 like kinase 1 also known as ATG1 protein              (56) Protein kinase superfamily(57) Autophagosmoe formation              (56) Up-regulated              (56) Diagnosis (58)

Gastric Cancer

Gastric cancer (GC) as the fifth most common malignancies in the world (59, 60) is the second primary cancer related lethal causing (-); in addition, about 800,000 cancer-related deaths are caused by gastric cancer each year worldwide announced by the World Health Organization (64). Due to late diagnosis, limited therapy options for most patients, the rate of 5-year survival is less than 20%. The most common type of stomach cancer is adenocarcinomas, which is divided into diffuse (undifferentiated) and intestinal (well-differentiated) and is about 90% of all; the rest are gastric lymphoma, gastrointestinal stromal tumors, and neuroendocrine tumors with about 3-5% of all malignant tumors of the stomach (65, 66) with high incident in  Asian countries (67). There are two types of gastric adenocarcinoma based upon anatomical location: cardia or proximal, and distal, non-cardia adenocarcinomas (68). The primarily carcinogen is known as Helicobacter pylori infection, which is considers as 60 to 70% of gastric cancer occurrence in the world. Studies showed that eradication of H.pylori has been successful in reducing gastric cancer risk (3). The prognosis is still not sufficient even though its incident has been declined in recent years in the United States. Regular methods for early detection and screening stomach cancer are ultrasonography, endoscopic sonography, double contrast radiologic method, computed tomography (CT), or magnetic resonance imaging (-). Some protein markers related to gastric cancer are tabulated in table 2.

Table 2.

The summarized protein markers specific to gastric cancer, the MeSH terms (NBCI Databases) of these markers are mentioned.

Disease Protein name Category Function Condition Application
Gastric carcinoma (72), Adenomas(73) Gastrokines :Gastrokine 1 (GKN1)           (74, 75) also known Antrum mucosal protein (AMP)-18     (76) GKN2 (77), GKN3(78) Family of stomach-specific proteins  (79) Growth inhibition           (75), Mucosal protection (72) Down-regulated   (80) Early detection(81)
Gastric carcinoma(82) Pepsinogen C (PGC)(83)also known as Gastricsin Belongs to the peptidase family A1 Aspartic proteinase(84) Low expression(85) Early detection(83)
Gastric carcinoma(86) IPO-38 antigen(87, 88), also known as N-L116 antigen Belongs to H2B histone(87) Proliferatingmarker(87) Up-regulated(88) Early detection(87)

Colon Cancer

Colorectal cancer is the second cause of cancer death with 30% inheritance base (89); in fact, half of all patients diagnosed with this invasive cancer would ultimately die (90, 91). Therefore, the prognosis is highly linked to early diagnosis; it leads to a five-year survival post-operation of over 80% while in the advanced conditions the five-year survival decreases to 40% (92).

Adenocarcinoma is the most frequent type of colon cancer (93), the rest includes gastrointestinal carcinoid tumors, gastrointestinal stromal tumors, primary colorectal lymphoma, leiomyosarcoma, melanoma and squamous cell carcinoma (-). Rectal bleeding, diarrhea, weight loss, abdominal pain, and anemia are considered to be the common symptoms of this malignancy (98). Risk factors that are accompanied with this disease are smoking and red meat consumption. On the other hand, the risk of colon cancer can be reduced by Aspirin (99, 100), consuming vegetables, grains, and fruit (101). Mainly colorectal cancer detection is divided into two category: 1) guaiac fecal occult blood testing (gFOBT), fecal immunochemical test (FIT) and testing stool for exfoliated DNA (sDNA) that is, stool test; and 2) Detecting lesions by methods including flexible sigmoidoscopy (FSIG), colonoscopy (CSPY), double-contrast barium enema (DCBE), computed tomography colonography (CTC), and colonoscopy (102). In addition, colonoscopy is an aggressive approach with approximately 90% effectiveness and not being able to detect all colonic polyps(103). These applications by being costly and invasive, causing possible bleeding or infection, bowel irritating, and possessing low specificity render to search for safer techniques providing minimum invasion, high performance, high degree of sensitivity and specificity, and inexpensive. In table 3, some of the putative protein markers of colorectal cancer are included (table3).

Table 3.

The putative tumor markers related to colon cancer.

Disease Protein name Category Function Condition Application
Colorectalcarcinoma  Proteasome subunit beta type 7 (PSB7)  (104) Cytoplasmic and nuclear protein(105) Integral to cellular proteolytic degradation capability(106) Up-regulated(105) Early detection(105)
 Colorectal adenocarcinoma(107 Colon cancer-specificantigens :
(CCSA)-2(108)
(CCSA)-3(109)
(CCSA)-4(110)
(CCSA)-5    (111)
Nuclear matrix proteins  (NMPs) (102)  Structural role in nuclear(109) Up-regulated(108) Early detection(112)

Discussion

Biomarkers are efficient diagnostic agents for detection and even early detection of diseases, especially malignant diseases (9,-). Biomarkers are diverse biochemical components such as DNA sequence, RNAs, proteins, and metabolites (116). The final goal of biomarker application is to introduce a sensitive and specific indicator for a certain kind of disease. For many kinds of diseases, this strategy is offered. For instance, prostate specific antigen (PSA) is a regular diagnostic biomarker for prostate cancer (117). Unfortunately, there is no specific biomarker for many kinds of malignant diseases (118,119). Therefore, intensive efforts for introducing a combination of various biomarkers (biomarker panel) are in progress (120,121). Biomarker panel study has been started to gain great attention in recent years (122). A single biomarker cannot be examined as accurate diagnosis tool due to cancer diversity, since several of them are reported in other diseases. Thus, the specificity and sensitivity of the evaluation can decrease. In a new approach combination of markers are applied to provide the efficient accuracy while a single of them is inadequate. These selected tumor markers should bring out less commonness between each cancers and other diseases especially cancer of same histological origin. In this study, these markers were selected based on most reported as a preferentially expressed marker in definite cancer during recent years. Each of these markers is chosen as independent marker, but it is necessary to combine and present them as a set of markers. Since these biomarkers may be related to other cancers, an accurate clinical examination can be a useful tool in distinguishing between different types of malignancies. Furthermore, the selected protein markers for the panel study are those that are observed in neoplasm with less histological origin. For example, some of these markers are seen in both esophageal and gastric cancers, so they were omitted for this research. Several proteins reported as candidate biomarkers for these cancers; nevertheless, not all represent the accurate diagnosis factors since they are common with other cancers. For clinical applications, more efforts should be done to managing these findings. In table 1, three markers related to esophagus cancer were achieved among them, (MAP3K3/MEKK3) over-expression plays a significant role in tumorigenesis (123).

This biomarker is also reported in other malignancies such as breast cancer and hepatocellular carcinoma (HCC) (124,125). The other marker, Jumonji/Arid1b (Jarid1b) protein modulates cell growth and its function is highly increased in esophageal, breast, melanoma, and prostate cancer (53, -). Protein marker, UNC-51 like kinase 1 can be also a specific diagnosis marker for esophageal neoplasm              (56). As it is mentioned in table 2, gastrokine family has an explicit role in gastric malignancy (-). Among these GKNs, gastrokin 3 is recently discovered (131). GKN1 also called antrum mucosal protein (AMP)-18     (76) is one of the most putative biomarkers (132). Its low expression can be concluded in onset of malignancy. Moreover, while in its presence in Helicobacter pylori-negative patients reported normal, it is decreased in Helicobacter pylori-positive patients (133). In addition, pepsinogen C plays a prominent role in gastric cancer (134). This marker is also reported to be an important marker in different gastric diseases. Its expression in superficial gastritis is completely lost while in other diseases such as gastric ulcer or erosion was reduced to 80%. On the other hand, this reduction is about 11% in gastric cancer (85).

Additionally, it is a known marker for breast cancer, and prostate cancer (135). IPO-38 that is expressed frequently through most stages of the cell cycle except during mitosis is another novel gastric tumor marker. It is also known as N-L116 antigen in many databases (136), and assigned for ameloblastomas malignancy (137). In addition, lots of highly reported gastric biomarkers such asReg IV (138, 139), serum amyloid A (SAA) (140, 141), Adipocytokine (142, 143),  sphingosine kinase 1 (SK1) (144, 145), and SM22 (146, 147) were observed during this review, but all of them were correlated with colon cancer as well. Thereby, in this study they are not considered as definite tumor marker due to having the same histological bases. As it is shown in table3, colon cancer biomarkers are introduced as specific markers including PSB7, colon-cancer specific antigen family. Colon cancer specific antigens including (CCSA)-2 (108), (CCSA)-3 (109),(CCSA)-4 (110), and (CCSA)-5        (111) are assigned with high sensitivity and specificity that are detectable in serum with high quantity. These potential markers are reported by many proteomic analyses (108, 109). Among all of these evaluated biomarkers, solely colon cancer biomarkers showed definite specificity comparing with esophageal cancer and gastric cancer. Most discovered tumor markers are not appropriate as standard clinical methods (148); a selected group of them could be useful as a feasible tool for clinical approaches. As it is reported in numerous researches, there are hundreds of nonspecific biomarkers for digestive system cancers. However, a small number of these proteins are merely observed in the nature of these cancers. More follow-up evaluations are required to lessen limitations in selective biomarker applications in clinical approaches. These verifications can be achieved via clinical examinations researches. Large-scale studies such as advanced proteomics can introduce new relevant biomarkers for achieving efficient panel (149). It can be concluded that non-invasive methods such as proteomics provides a great number of biomarkers (-) that can be managed by clinical approaches for efficient prognosis and diagnostic purposes in a format of biomarker panel. It seems that biomarker panels play an important role in cancer management especially in diagnostic procedures in near future.

Conclusion

Nemours protein biomarkers are introduced for esophageal, gastric and colon cancers. It seems that each of them has no adequate potential for clinical application due to insufficient specificity and sensitivity. This valuable source of biomarkers can be considered for formation of biomarker panels, which are useful and precious tools in cancer management. In general, biomarker analysis and clinical examination are two important activities that can lead to introducing novel biomarker panel for accurate prognosis, diagnosis, and patients follow up.

Acknowledgment

This article is derived from project code number 159-1312 entitled “The evaluation of protein panel markers of esophagus, gastric, and colon cancer: a proteomic approach” leaded by Mona Zamanian- Azodi, PhD student of Applied Proteomics.

References

  • 1.Dalilan S, Tavirani MR, Nabiuni M, Heidari-Keshel S, Azodi MZ, Zali H. Aqueous Extract of Lavender angustifolia Inhibits Lymphocytes Proliferation of Hodgkin’s Lymphoma Patients. Iran J Cancer Prev . 2013;6:201–8. [PMC free article] [PubMed] [Google Scholar]
  • 2.Tavirani M, Zali H, Nasiri S, Shokrgozar M. Human calprotectin, a potent anticancer with minimal side effect. Iran J Cancer Prev. 2009;2:183–88. [Google Scholar]
  • 3.Cho WC. Proteomics in translational cancer research: biomarker discovery for clinical applications. Expert Rev Proteomics. 2014;11:131–33. doi: 10.1586/14789450.2014.899908. [DOI] [PubMed] [Google Scholar]
  • 4.Nia YZ, Majd HA, Azodi M, Khayyer N. Using partitioning and non-partitioning clustering algorithms for included proteins sequences in esophagus, stomach and colon cancer. Journal of Paramedical Sciences. 2011;2:9–16. [Google Scholar]
  • 5.Wouters MW, Gooiker GA, van Sandick JW, Tollenaar RA. The volume‐outcome relation in the surgical treatment of esophageal cancer. Cancer. 2012;118:1754–63. doi: 10.1002/cncr.26383. [DOI] [PubMed] [Google Scholar]
  • 6.Sgourakis G, Gockel I, Karaliotas C, Moehler M, Schimanski CC, Schmidberger H, et al. Survival after chemotherapy and/or radiotherapy versus self-expanding metal stent insertion in the setting of inoperable esophageal cancer: a case-control study. BMC Cancer. 2012;12 doi: 10.1186/1471-2407-12-70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Polanski M, Anderson NL. A list of candidate cancer biomarkers for targeted proteomics. Biomark Insights. 2007;1:1–48. [PMC free article] [PubMed] [Google Scholar]
  • 8.Wang Q, Chaerkady R, Wu J, Hwang HJ, Papadopoulos N, Kopelovich L, et al. Mutant proteins as cancer-specific biomarkers. PNAS. 2011;108:2444–49. doi: 10.1073/pnas.1019203108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Nafar M, Kalantari S, Samavat S, Rezaei-Tavirani M, Rutishuser D, Zubarev RA. The Novel Diagnostic Biomarkers for Focal Segmental Glomerulosclerosis. ISRN nephrology. doi: 10.1155/2014/574261. PNAS [In press] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Kalantari S, Rutishauser D, Samavat S, Nafar M, Mahmudieh L, Rezaei-Tavirani M, et al. Urinary Prognostic Biomarkers and Classification of IgA Nephropathy by High Resolution Mass Spectrometry Coupled with Liquid Chromatography. PloSone. 2013;8:808–30. doi: 10.1371/journal.pone.0080830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Shi J LJ, Liao L, Guo Y, Wang H, Hu W, Hu T. Identification of candidate serum biomarkers for small cell lung cancer by proteomics analysis. Minerva Med. 2014;105:37–47. [PubMed] [Google Scholar]
  • 12.Ludwig JA, Weinstein JN. Biomarkers in cancer staging, prognosis and treatment selection. Nat Rev Cancer. 2005;5:845–56. doi: 10.1038/nrc1739. [DOI] [PubMed] [Google Scholar]
  • 13.Pepe MS, Etzioni R, Feng Z, Potter JD, Thompson ML, Thornquist M, et al. Phases of biomarker development for early detection of cancer. JNCI. 2001;93:1054–61. doi: 10.1093/jnci/93.14.1054. [DOI] [PubMed] [Google Scholar]
  • 14.Duffy M, Lamerz R, Haglund C, Nicolini A, Kalousova M, Holubec L, et al. Tumor markers in colorectal cancer, gastric cancer and gastrointestinal stromal cancers: European group on tumor markers 2014 guidelines update. Int J Cancer. 2014;134:2513–22. doi: 10.1002/ijc.28384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Pooladi M, Rezaei-Tavirani M, Hashemi M, Hesami-Tackallou S, Khaghani-Razi-Abad S, Moradi A, et al. The Study of “Dihydropyrimidinase Related Proteins (DRPs)” Expression Changes Influence in Malignant Astrocytoma Brain Tumor. Iran J Cancer Prev. 2014;7:130–33. [PMC free article] [PubMed] [Google Scholar]
  • 16.Srinivas PR, Verma M, Zhao Y, Srivastava S. Proteomics for cancer biomarker discovery. Clin chem. 2002;48:1160–69. [PubMed] [Google Scholar]
  • 17.Sawyers CL. The cancer biomarker problem. Nature. 2008;452:548–52. doi: 10.1038/nature06913. [DOI] [PubMed] [Google Scholar]
  • 18.Rusling JF, Kumar CV, Gutkind JS, Patel V. Measurement of biomarker proteins for point-of-care early detection and monitoring of cancer. Analyst. 2010;135:2496–11. doi: 10.1039/c0an00204f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Simpson RJ, Dorow DS. Cancer proteomics: from signaling networks to tumor markers. Trends Biochem Sci. 2001;19:40–48. doi: 10.1016/S0167-7799(01)01801-7. [DOI] [PubMed] [Google Scholar]
  • 20.Ryu J-W, Kim H-J, Lee Y-S, Myong N-H, Hwang C-H, Lee G-S, et al. The proteomics approach to find biomarkers in gastric cancer. J Korean Med Sci. 2003;18 doi: 10.3346/jkms.2003.18.4.505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Alaiya AA, Franzén B, Auer G, Linder S. Cancer proteomics: from identification of novel markers to creation of artifical learning models for tumor classification. Electrophoresis. 2000;21:1210–17. doi: 10.1002/(SICI)1522-2683(20000401)21:6<1210::AID-ELPS1210>3.0.CO;2-S. [DOI] [PubMed] [Google Scholar]
  • 22.Rai AJ, Chan DW. Cancer proteomics: serum diagnostics for tumor marker discovery. Ann N Y Acad Sci. 2004;1022:286–94. doi: 10.1196/annals.1318.044. [DOI] [PubMed] [Google Scholar]
  • 23.Firpo MA, Boucher KM, Mulvihill SJ. Prospects for developing an accurate diagnostic biomarker panel for low prevalence cancers. Theor Biol Med Model. 2014;11 doi: 10.1186/1742-4682-11-34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Chung L, Moore K, Phillips L, Boyle FM, Marsh DJ, Baxter RC. Novel serum protein biomarker panel revealed by mass spectrometry and its prognostic value in breast cancer. Breast Cancer Res Treat. 2014;16 doi: 10.1186/bcr3676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Wulfkuhle JD, Liotta LA, Petricoin EF. Proteomic applications for the early detection of cancer. Nat Rev Cancer. 2003;3:267–75. doi: 10.1038/nrc1043. [DOI] [PubMed] [Google Scholar]
  • 26.Zamanian-Azodi M, Vafaee R, Azodi T, Omidi R, Gilanchi S, Azizi-Jalilian F, et al. Molecular approaches in obesity studies. Gastroenterol Hepatol Bed Bench. 2013;6:S23–31. [PMC free article] [PubMed] [Google Scholar]
  • 27.Zamanian-Azodi M, Rezaei-Tavirani M, Mortazavian A, Vafaee R, Rezaei-Tavirani M, Zali H, et al. Application of Proteomics in Cancer Study. American Journal of Cancer Science. 2013;2:116–34. [Google Scholar]
  • 28.Jafari M, Mirzaie M, Sadeghi M, Marashi S-A, Rezaei-Tavirani M. Exploring biological processes involved in embryonic stem cell differentiation by analyzing proteomic data. Biochim Biophys Acta. 2013;1834:1063–69. doi: 10.1016/j.bbapap.2013.02.003. [DOI] [PubMed] [Google Scholar]
  • 29.Kusiak RA, Ritchie AC, Springer J, Muller J. Mortality from stomach cancer in Ontario miners. Br J Ind Med. 1993;50:117–26. doi: 10.1136/oem.50.2.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Ricci-Vitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M, Peschle C, et al. Identification and expansion of human colon-cancer-initiating cells. Nature. 2007;445:111–15. doi: 10.1038/nature05384. [DOI] [PubMed] [Google Scholar]
  • 31.Zali H, Ahmadi G, Bakhshandeh R, Rezaei-Tavirani M. Proteomic analysis of gene expression during human esophagus cancer. Journal of Paramedical Sciences. 2011;2:37–44. [Google Scholar]
  • 32.Enzinger PC, Mayer RJ. Esophageal cancer. N Engl J Med. 2003;349:2241–52. doi: 10.1056/NEJMra035010. [DOI] [PubMed] [Google Scholar]
  • 33.Agrawal N, Jiao Y, Bettegowda C, Hutfless SM, Wang Y, David S, et al. Comparative Genomic Analysis of Esophageal Adenocarcinoma and Squamous Cell Carcinoma. Cancer Discov. 2012;2:899–905. doi: 10.1158/2159-8290.CD-12-0189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Shakhatreh MH, Duan Z, Avila N, Naik AD, Kramer JR, Hinojosa-Lindsey M, et al. Risk of upper gastrointestinal cancers in patients with gastroesophageal reflux disease following a negative screening endoscopy. Clin Gastroenterol Hepatol . doi: 10.1016/j.cgh.2014.06.029. [In press] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Yoon HH, Shi Q, Sukov WR, Wiktor AE, Khan M, Sattler CA, et al. Association of HER2/ErbB2 expression and gene amplification with pathologic features and prognosis in esophageal adenocarcinomas. Clin Cancer Res. 2012;18:546–54. doi: 10.1158/1078-0432.CCR-11-2272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Madhusudhan C, Saluja SS, Pal S, Ahuja V, Saran P, Dash NR, et al. Palliative stenting for relief of dysphagia in patients with inoperable esophageal cancer: impact on quality of life. Dis Esophagus. 2009;22:331–36. doi: 10.1111/j.1442-2050.2008.00906.x. [DOI] [PubMed] [Google Scholar]
  • 37.Daly JM, Fry WA, Little AG, Winchester DP, McKee RF, Stewart AK, et al. Esophageal cancer: results of an American College of Surgeons patient care evaluation study. JCPSP. 2000;190:562–72. doi: 10.1016/s1072-7515(00)00238-6. [DOI] [PubMed] [Google Scholar]
  • 38.Layke JC, Lopez PP. Esophageal cancer: a review and update. AmFam Physician. 2006;73:2187–94. [PubMed] [Google Scholar]
  • 39.Mao W-M, Zheng W-H, Ling Z-Q. Epidemiologic risk factors for esophageal cancer development. Asian Pac J Cancer Prev. 2011;12:2461–66. [PubMed] [Google Scholar]
  • 40.Oze I, Matsuo K, Wakai K, Nagata C, Mizoue T, Tanaka K, et al. Alcohol drinking and esophageal cancer risk: an evaluation based on a systematic review of epidemiologic evidence among the Japanese population. J ClinOncol Res. 2011;41:677–92. doi: 10.1093/jjco/hyr026. [DOI] [PubMed] [Google Scholar]
  • 41.Salehi M, Moradi‐Lakeh M, Salehi MH, Nojomi M, Kolahdooz F. Meat, fish, and esophageal cancer risk: a systematic review and dose‐response meta‐analysis. Nutr Rev. 2013;71:257–67. doi: 10.1111/nure.12028. [DOI] [PubMed] [Google Scholar]
  • 42.Shaheen N, Ransohoff DF. Gastroesophageal reflux, Barrett esophagus, and esophageal cancer: scientific review. JAMA. 2002;287:1972–81. doi: 10.1001/jama.287.15.1972. [DOI] [PubMed] [Google Scholar]
  • 43.Cerfolio RJ, Bryant AS, Ohja B, Bartolucci AA, Eloubeidi MA. The accuracy of endoscopic ultrasonography with fine-needle aspiration, integrated positron emission tomography with computed tomography, and computed tomography in restaging patients with esophageal cancer after neoadjuvantchemoradiotherapy. J Thorac Cardiovasc Surg. 2005;129:1232–41. doi: 10.1016/j.jtcvs.2004.12.042. [DOI] [PubMed] [Google Scholar]
  • 44.Jäckle S, Gladkova N, Feldchtein F, Terentieva A, Brand B, Gelikonov G, et al. In vivo endoscopic optical coherence tomography of esophagitis, Barrett's esophagus, and adenocarcinoma of the esophagus. Endoscopy. 2000;32:750–55. doi: 10.1055/s-2000-7705. [DOI] [PubMed] [Google Scholar]
  • 45.Komukai S, Nishimaki T, Watanabe H, Ajioka Y, Suzuki T, Hatakeyama K. Significance of immunohistochemically demonstrated micrometastases to lymph nodes in esophageal cancer with histologically negative nodes. Surgery. 2000;127:40–46. doi: 10.1067/msy.2000.102754. [DOI] [PubMed] [Google Scholar]
  • 46.Wu S, Chen M, Wei L, Chen Z. Embedded Cervical Esophagogastrostomy: A Simple and Convenient Method Using a Circular Stapler After Esophagectomy for Esophageal Carcinomas. Ann Surg Oncol. 2013;20:2984–90. doi: 10.1245/s10434-013-2991-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Smolinski KN, Abraham JM, Souza RF, Yin J, Wang S, Xu Y, et al. Activation of the Esophagin Promoter during Esophageal Epithelial Cell Differentiation. Genomics. 2002;79:875–80. doi: 10.1006/geno.2002.6775. [DOI] [PubMed] [Google Scholar]
  • 48.Hasan R, Sharma R, Saraya A, Chattopadhyay TK, DattaGupta S, Walfish PG, et al. Mitogen activated protein kinase kinasekinase 3 (MAP3K3/MEKK3) overexpression is an early event in esophageal tumorigenesis and is a predictor of poor disease prognosis. BMC Cancer. 2014;14 doi: 10.1186/1471-2407-14-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Ogawa R, Ishiguro H, Kuwabara Y, Kimura M, Mitsui A, Katada T, et al. Expression profiling of micro-RNAs in human esophageal squamous cell carcinoma using RT-PCR. Med Mol Morphol. 2009;42:102–109. doi: 10.1007/s00795-009-0443-1. [DOI] [PubMed] [Google Scholar]
  • 50.Shi Y. Histone lysine demethylases: emerging roles in development, physiology and disease. Nature Rev Genet. 2007;8:829–33. doi: 10.1038/nrg2218. [DOI] [PubMed] [Google Scholar]
  • 51.Kano Y, Konno M, Ohta K, Haraguchi N, Nishikawa S, Kagawa Y, et al. Jumonji/Arid1b (Jarid1b) protein modulates human esophageal cancer cell growth. Mol Clin Oncol. 2013;1:753–57. doi: 10.3892/mco.2013.127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Matias NMR. Exploring the role of the Jumonji/ARID1 family of proteins during embryonic stem cell differentiation and X-chromosome inactivation. 2010;1:753–57. [Google Scholar]
  • 53.Kano Y, Konno M, Kawamoto K, Tamari K, Hayashi K, Fukusumi T, et al. Novel drug discovery system for cancer stem cells in human squamous cell carcinoma of the esophagus. Oncol Rep. 2014;31:1133–38. doi: 10.3892/or.2013.2952. [DOI] [PubMed] [Google Scholar]
  • 54.Roesch AO, Herlyn M. Compositions containing JARID1B inhibitors and methods for treating cancer. Available from: http://www.google.com/patents/CA2770307A1?cl=en.
  • 55.Nakamura Y, Hamamoto R, Tsunoda T. Jarid1b for target gene of cancer therapy and diagnosis. Available from: http://www.google.com/patents/EP2398901A1?cl=en.
  • 56.Jiang S, Li Y, Zhu YH, Wu XQ, Tang J, Li Z, et al. Intensive expression of UNC‐51‐like kinase 1 is a novel biomarker of poor prognosis in patients with esophageal squamous cell carcinoma. Cancer Sci. 2011;102:1568–75. doi: 10.1111/j.1349-7006.2011.01964.x. [DOI] [PubMed] [Google Scholar]
  • 57.Ogura K-i, Okada T, Mitani S, Gengyo-Ando K, Baillie DL, Kohara Y, et al. Protein phosphatase 2A cooperates with the autophagy-related kinase UNC-51 to regulate axon guidance in Caenorhabditiselegans. Development. 2010;137:1657–67. doi: 10.1242/dev.050708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Jiang L, Duan B-S, Huang J-X, Jiao X, Zhu X-W, Sheng H-H, et al. Association of the expression of unc-51-Like kinase 1 with lymph node metastasis and survival in patients with esophageal squamous cell carcinoma. Int J Clin Exp Med. 2014;7 [PMC free article] [PubMed] [Google Scholar]
  • 59.Zali H, Rezaei-Tavirani M, Vafaee R, Rezaei-Tavirani M. Gastric cardia adenocarcinoma pathway analysis. Gastroenterol Hepatol Bed Bench. 2013;6:S11–18. [PMC free article] [PubMed] [Google Scholar]
  • 60.Fock KM. Review article: the epidemiology and prevention of gastric cancer. Aliment Pharmacol Ther. 2014;40:250–60. doi: 10.1111/apt.12814. [DOI] [PubMed] [Google Scholar]
  • 61.Khatib H, Rezaei-Tavirani M, Keshel SH, Azodi MZ, Omidi R, Biglarian M, et al. Flow cytometry analysis of Rosa Damascena effects on gastric cancer cell line (MKN45) Iran J Cancer Prev. 2013;6:30–36. [Google Scholar]
  • 62.Shokrgozar M, Zali H, RezaeiTavirani M. Evaluation of proliferation inhibition effect of human calprotectin on human gastric cancer cell line (AGS) in vitro. Yakhteh. 2007;4:254–68. [Google Scholar]
  • 63.Zali H, Zamanian-Azodi M, Shokrgozar MA, Rezaei-Tavirani M. Cytotoxic effects of human calprotectin on gastric cancer cell line is attenuated by etoposide. Gastroenterol Hepatol Bed Bench. 2012;5:132–38. [PMC free article] [PubMed] [Google Scholar]
  • 64.Liu W, Yang Q, Liu B, Zhu Z. Serum proteomics for gastric cancer. Clinica Chimica Acta. 2014;431:179–84. doi: 10.1016/j.cca.2014.02.001. [DOI] [PubMed] [Google Scholar]
  • 65.Hendricks J. Malignant tumors of the stomach. Surgclin North Am. 1986;66:683–93. doi: 10.1016/s0039-6109(16)43981-2. [DOI] [PubMed] [Google Scholar]
  • 66.Okines A, Verheij M, Allum W, Cunningham D, Cervantes A. Gastric cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2010;21:v50–v54. doi: 10.1093/annonc/mdq164. [DOI] [PubMed] [Google Scholar]
  • 67.He Q-Y, Cheung YH, Leung SY, Yuen ST, Chu K-M, Chiu J-F. Diverse proteomic alterations in gastric adenocarcinoma. Proteomics. 2004;4:3276–87. doi: 10.1002/pmic.200300916. [DOI] [PubMed] [Google Scholar]
  • 68.Ghimire P, Wu G-Y, Zhu L. Primary gastrointestinal lymphoma. World J Gastroenterol. 2011;17:697–707. doi: 10.3748/wjg.v17.i6.697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Goto R, Arai K, Kitada H, Ogoshi K, Hamashima C. Labor Resource Use for Endoscopic Gastric Cancer Screening in Japanese Primary Care Settings: A Work Sampling Study. PloSone. 2014;9:1–6. doi: 10.1371/journal.pone.0088113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Hisamichi S. Screening for gastric cancer. World J Surg. 1989;13:31–37. doi: 10.1007/BF01671151. [DOI] [PubMed] [Google Scholar]
  • 71.Stell D, Carter C, Stewart I, Anderson J. Prospective comparison of laparoscopy, ultrasonography and computed tomography in the staging of gastric cancer. Br J Surg. 1996;83:1260–62. [PubMed] [Google Scholar]
  • 72.Oien KA, McGregor F, Butler S, Ferrier RK, Downie I, Bryce S, et al. Gastrokine 1 is abundantly and specifically expressed in superficial gastric epithelium, down‐regulated in gastric carcinoma, and shows high evolutionary conservation. J Pathol. 2004;203:789–97. doi: 10.1002/path.1583. [DOI] [PubMed] [Google Scholar]
  • 73.Yoon JH, Song JH, Zhang C, Jin M, Kang YH, Nam SW, et al. Inactivation of the Gastrokine 1 gene in gastric adenomas and carcinomas. J Pathol. 2011;223:618–25. doi: 10.1002/path.2838. [DOI] [PubMed] [Google Scholar]
  • 74.Wu J-Y, Cheng C-C, Wang J-Y, Wu D-C, Hsieh J-S, Lee S-C, et al. Discovery of tumor markers for gastric cancer by proteomics. PloSone. 2014;9 doi: 10.1371/journal.pone.0084158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Yan GR, Xu SH, Tan ZL, Yin XF, He QY. Proteomics characterization of gastrokine 1‐induced growth inhibition of gastric cancer cells. Proteomics. 2011;11:3657–64. doi: 10.1002/pmic.201100215. [DOI] [PubMed] [Google Scholar]
  • 76.He QY, Cheung YH, Leung SY, Yuen ST, Chu KM, Chiu JF. Diverse proteomic alterations in gastric adenocarcinoma. Proteomics. 2004;4:3276–87. doi: 10.1002/pmic.200300916. [DOI] [PubMed] [Google Scholar]
  • 77.Baus-Loncar M, Lubka M, Pusch CM, Otto WR, Poulsom R, Blin N. Cytokine regulation of the trefoil factor family binding protein GKN2 (GDDR/TFIZ1/blottin) in human gastrointestinal epithelial cells. Cell Physiol Biochem. 2007;20:193–204. doi: 10.1159/000104166. [DOI] [PubMed] [Google Scholar]
  • 78.Menheniott TR, Kurklu B, Giraud AS. Gastrokines: stomach-specific proteins with putative homeostatic and tumor suppressor roles. Am J PhysiolGastrointest Liver Physiol. 2013;304:109–21. doi: 10.1152/ajpgi.00374.2012. [DOI] [PubMed] [Google Scholar]
  • 79.Rippa E, La Monica G, Allocca R, Romano MF, De Palma M, Arcari P. Overexpression of gastrokine 1 in gastric cancer cells induces Fas‐mediated apoptosis. J Cell Physiol. 2011;226:2571–8. doi: 10.1002/jcp.22601. [DOI] [PubMed] [Google Scholar]
  • 80.Martin G, Wex T, Treiber G, Malfertheiner P, Nardone G. Low‐dose aspirin reduces the gene expression of gastrokine‐1 in the antral mucosa of healthy subjects. Aliment pharmacol Ther. 2008;28:782–88. doi: 10.1111/j.1365-2036.2008.03793.x. [DOI] [PubMed] [Google Scholar]
  • 81.Cui J, Chen Y, Chou W-C, Sun L, Chen L, Suo J, et al. An integrated transcriptomic and computational analysis for biomarker identification in gastric cancer. Nucleic Acids Res. 2011;39:1197–207. doi: 10.1093/nar/gkq960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Abnet CC. Current and ongoing research projects related to gastric cancer prevention: perspective of the United States National Cancer Institute. Helicobacter pylori eradication as strategy for preventing gastric cancer. Lyon, France: International Agency for Research on Cancer; 2014. p. 136. [Google Scholar]
  • 83.Melle C, Ernst G, Schimmel B, Bleul A, Kaufmann R, Hommann M, et al. Characterization of pepsinogen C as a potential biomarker for gastric cancer using a histo-proteomic approach. Journal Proteome Rev. 2005;4:1799–804. doi: 10.1021/pr050123o. [DOI] [PubMed] [Google Scholar]
  • 84.Dame JB, Reddy GR, Yowell CA, Dunn BM, Kay J, Berry C. Sequence, expression and modeled structure of an aspartic proteinase from the human malaria parasitePlasmodium falciparum. Mol Biochem Parasit. 1994;64:177–90. doi: 10.1016/0166-6851(94)90024-8. [DOI] [PubMed] [Google Scholar]
  • 85.Li H-m, Ning P-f, Yuan Y. Contrastive study of the tissue expression and serum concentration of pepsinogen C in gastric mucosa diseases. Chines J Cancer Res. 2006;18:8–11. [Google Scholar]
  • 86.Chen L, Su L, Li J, Zheng Y, Yu B, Yu Y, et al. Hypermethylated FAM5C and MYLK in serum as diagnosis and pre-warning markers for gastric cancer. Dis Markers. 2012;32(3):195–202. doi: 10.3233/DMA-2011-0877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Hao Y, Yu Y, Wang L, Yan M, Ji J, Qu Y, et al. IPO-38 is identified as a novel serum biomarker of gastric cancer based on clinical proteomics technology. J Proteome Res. 2008;7:3668–77. doi: 10.1021/pr700638k. [DOI] [PubMed] [Google Scholar]
  • 88.Lin L-L, Huang H-C, Juan H-F. Discovery of biomarkers for gastric cancer: a proteomics approach. J Proteomics. 2012;75:3081–97. doi: 10.1016/j.jprot.2012.03.046. [DOI] [PubMed] [Google Scholar]
  • 89.Jasperson KW, Tuohy TM, Neklason DW, Burt RW. Hereditary and familial colon cancer. Gastroenterology. 2010;138:2044–58. doi: 10.1053/j.gastro.2010.01.054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Rezaie-Tavirani M, Fayazfar S, Heydari-Keshel S, Rezaee MB, Zamanian-Azodi M, Rezaei-Tavirani M, et al. Effect of essential oil of Rosa Damascena on human colon cancer cell line SW742. Gastroenterol Hepatol Bed Bench. 2013;6:25–31. [PMC free article] [PubMed] [Google Scholar]
  • 91.Safaei A, Sobhi S, Rezaei-Tavirani M, Zali MR. Genomic and epigenetic instability in colorectal cancer. Iran J Cancer Prev. 2013;6:54–63. [PMC free article] [PubMed] [Google Scholar]
  • 92.Yu J-K, Chen Y-D, Zheng S. An integrated approach to the detection of colorectal cancer utilizing proteomics and bioinformatics. World J Gastroenterol. 2004;10:3127–31. doi: 10.3748/wjg.v10.i21.3127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Snover DC. Update on the serrated pathway to colorectal carcinoma. Hum Pathol. 2011;42:1–10. doi: 10.1016/j.humpath.2010.06.002. [DOI] [PubMed] [Google Scholar]
  • 94.Fan C-W, Changchien CR, Wang J-Y, Chen J-S, Hsu K-C, Tang R, et al. Primary colorectal lymphoma. Dis Colon Rectum. 2000;43:1277–82. doi: 10.1007/BF02237436. [DOI] [PubMed] [Google Scholar]
  • 95.Comer TP, Beahrs OH, Dockerty MB. Primary squamous cell carcinoma and adenoacanthoma of the colon. Cancer. 1971;28:1111–17. doi: 10.1002/1097-0142(1971)28:5<1111::aid-cncr2820280504>3.0.co;2-v. [DOI] [PubMed] [Google Scholar]
  • 96.Miettinen M, Sarlomo-Rikala M, Sobin LH, Lasota J. Gastrointestinal stromal tumors and leiomyosarcomas in the colon: a clinicopathologic, immunohistochemical, and molecular genetic study of 44 cases. Am Surg Pathol. 2000;24:1339–52. doi: 10.1097/00000478-200010000-00003. [DOI] [PubMed] [Google Scholar]
  • 97.Stinner B, Kisker O, Zielke A, Rothmund M. Surgical management for carcinoid tumors of small bowel, appendix, colon, and rectum. World J Surg. 1996;20:183–88. doi: 10.1007/s002689900028. [DOI] [PubMed] [Google Scholar]
  • 98.Astin M, Griffin T, Neal RD, Rose P, Hamilton W. The diagnostic value of symptoms for colorectal cancer in primary care: a systematic review. Br J Gen Pract. 2011;61:231–43. doi: 10.3399/bjgp11X572427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Nishihara R, Lochhead P, Kuchiba A, et al. ASpirin use and risk of colorectal cancer according to braf mutation status. JAMA. 2013;309:2563–71. doi: 10.1001/jama.2013.6599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Thun MJ, Jacobs EJ, Patrono C. The role of aspirin in cancer prevention. Nat Rev Clin Oncol. 2012;9:259–67. doi: 10.1038/nrclinonc.2011.199. [DOI] [PubMed] [Google Scholar]
  • 101.Trock B, Lanza E, Greenwald P. Dietary fiber, vegetables, and colon cancer: Critical review and meta-analyses of the epidemiologic evidence. J Natl Cancer Inst. 1990;82:650–61. doi: 10.1093/jnci/82.8.650. [DOI] [PubMed] [Google Scholar]
  • 102.Xue G, Wang X, Yang Y, Liu D, Cheng Y, Zhou J, et al. Colon Cancer-Specific Antigen-2 May Be Used as a Detecting and Prognostic Marker in Colorectal Cancer: A Preliminary Observation. PloSone. 2014;9 doi: 10.1371/journal.pone.0094252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Young PE, Womeldorph CM. Colonoscopy for colorectal cancer screening. J Cancer. 2013;4 doi: 10.7150/jca.5829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Shen H, Huang J, Pei H, Zeng S, Tao Y, Shen L, et al. Comparative proteomic study for profiling differentially expressed proteins between Chinese left‐and right‐sided colon cancers. Cancer Sci. 2013;104:135–41. doi: 10.1111/cas.12029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Rho J-h, Qin S, Wang JY, Roehrl MH. Proteomic expression analysis of surgical human colorectal cancer tissues: up-regulation of PSB7, PRDX1, and SRP9 and hypoxic adaptation in cancer. J Proteome Res. 2008;7:2959–72. doi: 10.1021/pr8000892. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Coux O, Tanaka K, Goldberg AL. Structure and functions of the 20S and 26S proteasomes. Ann Rev Biochem. 1996;65:801–47. doi: 10.1146/annurev.bi.65.070196.004101. [DOI] [PubMed] [Google Scholar]
  • 107.Knychalski B, Łukieńczuk T. The evaluation of diagnostic value of the tumor markers: CCSA-2 and CEA in colorectal cancer. Pol J Surg. 2012;84:86–92. doi: 10.2478/v10035-012-0014-3. [DOI] [PubMed] [Google Scholar]
  • 108.Leman ES, Schoen RE, Magheli A, Sokoll LJ, Chan DW, Getzenberg RH. Evaluation of colon cancer–specific antigen 2 as a potential serum marker for colorectal cancer. Clin. Cancer Res. 2008;14:1349–54. doi: 10.1158/1078-0432.CCR-07-4110. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 109.Leman ES, Schoen RE, Weissfeld JL, Cannon GW, Sokoll LJ, Chan DW, et al. Initial analyses of colon cancer–specific antigen (CCSA)-3 and CCSA-4 as colorectal cancer–associated serum markers. Cancer Res. 2007;67:5600–605. doi: 10.1158/0008-5472.CAN-07-0649. [DOI] [PubMed] [Google Scholar]
  • 110.Chen X, Ba Y, Ma L, Cai X, Yin Y, Wang K, et al. Characterization of microRNAs in serum: a novel class of biomarkers for diagnosis of cancer and other diseases. Cell Res. 2008;18:997–1006. doi: 10.1038/cr.2008.282. [DOI] [PubMed] [Google Scholar]
  • 111.Walgenbach‐Brunagel G, Burger B, Leman ES, Walgenbach KJ, Tolba R, Heukamp L, et al. The use of a colon cancer associated nuclear antigen CCSA‐2 for the blood based detection of colon cancer. J Cell Biochem. 2008;104:286–94. doi: 10.1002/jcb.21619. [DOI] [PubMed] [Google Scholar]
  • 112.Gupta AK, Brenner DE, Turgeon DK. Early Detection of Colon Cancer. Mol Diag Ther. 2008;12:77–85. doi: 10.1007/BF03256273. [DOI] [PubMed] [Google Scholar]
  • 113.Rezaei TM, Zali H, Rastegar JF, Heydari H, Hosseinzadeh SB, Daneshi MF, et al. Paper: Introducing Aldolase C as A Differentional Biomarker: A Proteomics Approach. Mol Diag Ther. 2008;12:77–85. [Google Scholar]
  • 114.Hasanzadeh H, Rezaie-Tavirani M, Seyyedi SS, Zali H, Keshel SH, Jadidi M, et al. Effect of ELF-EMF Exposure on Human Neuroblastoma Cell Line: a Proteomics Analysis. Iran J Cancer Prev. 2014;7:22–27. [PMC free article] [PubMed] [Google Scholar]
  • 115.De Vita F, Di Martino N, Fabozzi A, Laterza MM, Ventriglia J, Savastano B, et al. Clinical management of advanced gastric cancer: The role of new molecular drugs. World J Gastroenterol. 2014;20:14537–58. doi: 10.3748/wjg.v20.i40.14537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Babashah S, Sadeghizadeh M, Tavirani MR, Farivar S, Soleimani M. Aberrant microRNA expression and its implications in the pathogenesis of leukemias. Cell Oncol. 2012;35:317–34. doi: 10.1007/s13402-012-0095-3. [DOI] [PubMed] [Google Scholar]
  • 117.Partin AW, Catalona WJ, Southwick PC, Subong EN, Gasior GH, Chan DW. Analysis of percent free prostate-specific antigen (PSA) for prostate cancer detection: influence of total PSA, prostate volume, and age. Urology. 1996;48:55–61. doi: 10.1016/s0090-4295(96)00611-5. [DOI] [PubMed] [Google Scholar]
  • 118.Zamanian-Azodi M, Rezaie-Tavirani M, Heydari-Kashal S, Kalantari S, Dailian S, Zali H. Proteomics analysis of MKN45 cell line before and after treatment with Lavender aqueous extract. Gastroenterol Hepatol Bed Bench. 2012;5:35–42. [PMC free article] [PubMed] [Google Scholar]
  • 119.Maurya P, Meleady P, Dowling P, Clynes M. Proteomic approaches for serum biomarker discovery in cancer. Anticancer Res. 2007;27:1247–55. [PubMed] [Google Scholar]
  • 120.Urakami S, Shiina H, Enokida H, Kawakami T, Kawamoto K, Hirata H, et al. Combination analysis of hypermethylatedWnt-antagonist family genes as a novel epigenetic biomarker panel for bladder cancer detection. Clin Cancer Res. 2006;12:2109–16. doi: 10.1158/1078-0432.CCR-05-2468. [DOI] [PubMed] [Google Scholar]
  • 121.Ring BZ, Seitz RS, Beck R, Shasteen WJ, Tarr SM, Cheang MC, et al. Novel prognostic immunohistochemical biomarker panel for estrogen receptor–positive breast cancer. J Clin Oncol. 2006;24:3039–47. doi: 10.1200/JCO.2006.05.6564. [DOI] [PubMed] [Google Scholar]
  • 122.Nie S, Lo A, Wu J, Zhu J, Tan Z, Simeone DM, et al. Glycoprotein Biomarker Panel for Pancreatic Cancer Discovered by Quantitative Proteomics Analysis. J proteome Res. 2014;13:1873–84. doi: 10.1021/pr400967x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Kumar A, Chatopadhyay T, Raziuddin M, Ralhan R. Discovery of deregulation of zinc homeostasis and its associated genes in esophageal squamous cell carcinoma using cDNA microarray. Int J Cancer. 2007;120:230–42. doi: 10.1002/ijc.22246. [DOI] [PubMed] [Google Scholar]
  • 124.Zhang HA. MAP3K3/MEKK3 Is an amplified and overexpressed novel oncogene in breast cancer. J Clin Exp Pathol. 2012;2:25–32. [Google Scholar]
  • 125.Burchard J, Zhang C, Liu AM, Poon RT, Lee NP, Wong KF, et al. MicroRNA‐122 as a regulator of mitochondrial metabolic gene network in hepatocellular carcinoma. Mol Syst Biol. 2010;6 doi: 10.1038/msb.2010.58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Xiang Y, Zhu Z, Han G, Ye X, Xu B, Peng Z, et al. JARID1B is a histone H3 lysine 4 demethylase up-regulated in prostate cancer. ProcNatlAcadSci U S A. 2007;104:19226–31. doi: 10.1073/pnas.0700735104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Mitra D, Das PM, Huynh FC, Jones FE. Jumonji/ARID1 B (JARID1B) protein promotes breast tumor cell cycle progression through epigenetic repression of microRNA let-7e. J Biol Chem. 2011;286:40531–35. doi: 10.1074/jbc.M111.304865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Kuzbicki L, Lange D, Straczynska-Niemiec A, Chwirot BW. JARID1B expression in human melanoma and benign melanocytic skin lesions. Melanoma Res. 2013;23:8–12. doi: 10.1097/CMR.0b013e32835d5d6f. [DOI] [PubMed] [Google Scholar]
  • 129.Dai J, Zhang N, Wang J, Chen M, Chen J. Gastrokine-2 is downregulated in gastric cancer and its restoration suppresses gastric tumorigenesis and cancer metastasis. Tumor Biol. 2014;35:4199–207. doi: 10.1007/s13277-013-1550-0. [DOI] [PubMed] [Google Scholar]
  • 130.Lu F, Tempera I, Lee HT, DeWispelaere K, Lieberman PM. EBNA1 binding and epigenetic regulation of gastrokine tumor suppressor genes in gastric carcinoma cells. Virol J. 2014;11 doi: 10.1186/1743-422X-11-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Menheniott TR, Peterson AJ, O'Connor L, Lee KS, Kalantzis A, Kondova I, et al. A Novel<i>Gastrokine, Gkn3, Marks Gastric Atrophy and Shows Evidence of Adaptive Gene Loss in Humans. Gastroenterology. 2010;138:1823–35. doi: 10.1053/j.gastro.2010.01.050. [DOI] [PubMed] [Google Scholar]
  • 132.Altieri F, Stadio D, Stella C, Severino V, Sandomenico A, Minopoli G, et al. Anti-amyloidogenic property of human gastrokine 1. Biochimie. 2014;106:91–100. doi: 10.1016/j.biochi.2014.08.004. [DOI] [PubMed] [Google Scholar]
  • 133.Nardone G, Martin G, Rocco A, Rippa E, La Monica G, Caruso F, et al. Molecular expression of Gastrokine 1 in normal mucosa and in Helicobacter pylori-related preneoplastic and neoplastic gastric lesions. Cancer Biol Ther. 2008;7:1890–95. doi: 10.4161/cbt.7.12.6936. [DOI] [PubMed] [Google Scholar]
  • 134.Ning P-F, Liu H-J, Yuan Y. Dynamic expression of pepsinogen C in gastric cancer, precancerous lesions and Helicobacter pylori associated gastric diseases. World J Gastroenterol. 2005;11:2545–48. doi: 10.3748/wjg.v11.i17.2545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Balbín M, López-Otín C. Hormonal Regulation of the Human Pepsinogen C Gene in Breast Cancer Cells Identification of Cis-Acting Element Mediating Its Induction by Androgens, Glucocorticoids, and Progestrone. J Biol Chem. 1996;271:15175–81. doi: 10.1074/jbc.271.25.15175. [DOI] [PubMed] [Google Scholar]
  • 136.Thosaporn W, Iamaroon A, Pongsiriwet S, Ng K. A comparative study of epithelial cell proliferation between the odontogenickeratocyst, orthokeratinizedodontogenic cyst, dentigerous cyst, and ameloblastoma. Oral Dis. 2004;10:22–6. doi: 10.1046/j.1354-523x.2003.00974.x. [DOI] [PubMed] [Google Scholar]
  • 137.Amaral FR, Mateus GCP, Bonisson LA, Andrade BABd, Mesquita RA, Horta MCR, et al. Cell proliferation and apoptosis in ameloblastomas and keratocysticodontogenic tumors. Braz Dent J. 2012;23:91–96. doi: 10.1590/s0103-64402012000200001. [DOI] [PubMed] [Google Scholar]
  • 138.Violette S, Festor E, Pandrea‐Vasile I, Mitchell V, Adida C, Dussaulx E, et al. Reg IV, a new member of the regenerating gene family, is overexpressed in colorectal carcinomas. Intl J Cancer. 2003;103:185–93. doi: 10.1002/ijc.10788. [DOI] [PubMed] [Google Scholar]
  • 139.Mitani Y, Oue N, Matsumura S, Yoshida K, Noguchi T, Ito M, et al. Reg IV is a serum biomarker for gastric cancer patients and predicts response to 5-fluorouracil-based chemotherapy. Oncogene. 2007;26:4383–93. doi: 10.1038/sj.onc.1210215. [DOI] [PubMed] [Google Scholar]
  • 140.Michaeli A, Finci-Yeheskel Z, Dishon S, Linke RP, Levin M, Urieli-Shoval S. Serum amyloid A enhances plasminogen activation: implication for a role in colon cancer. BiochemBiophys Res Commun. 2008;368:368–73. doi: 10.1016/j.bbrc.2008.01.079. [DOI] [PubMed] [Google Scholar]
  • 141.Chan D-C, Chen C-J, Chu H-C, Chang W-K, Yu J-C, Chen Y-J, et al. Evaluation of serum amyloid A as a biomarker for gastric cancer. Ann Surg Oncol. 2007;14:84–93. doi: 10.1245/s10434-006-9091-z. [DOI] [PubMed] [Google Scholar]
  • 142.Nakajima TE, Yamada Y, Hamano T, Furuta K, Matsuda T, Fujita S, et al. Adipocytokines as new promising markers of colorectal tumors: adiponectin for colorectal adenoma, and resistin and visfatin for colorectal cancer. Cancer Sci. 2010;101:1286–91. doi: 10.1111/j.1349-7006.2010.01518.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Nakajima TE, Yamada Y, Hamano T, Furuta K, Gotoda T, Katai H, et al. Adipocytokine levels in gastric cancer patients: resistin and visfatin as biomarkers of gastric cancer. Am J Gastroenterol. 2009;44:685–90. doi: 10.1007/s00535-009-0063-5. [DOI] [PubMed] [Google Scholar]
  • 144.Kawamori T, Kaneshiro T, Okumura M, Maalouf S, Uflacker A, Bielawski J, et al. Role for sphingosinekinase 1 in colon carcinogenesis. FASEB J. 2009;23:405–14. doi: 10.1096/fj.08-117572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Li W, Yu C-P, Xia J-t, Zhang L, Weng G-X, Zheng H-q, et al. Sphingosine kinase 1 is associated with gastric cancer progression and poor survival of patients. Clin Cancer Res. 2009;15(4):1393–9. doi: 10.1158/1078-0432.CCR-08-1158. [DOI] [PubMed] [Google Scholar]
  • 146.Yeo M, Park HJ, Kim DK, Kim YB, Cheong JY, Lee KJ, et al. Loss of SM22 is a characteristic signature of colon carcinogenesis and its restoration suppresses colon tumorigenicity in vivo and in vitro. Cancer. 2010;116:2581–89. doi: 10.1002/cncr.25003. [DOI] [PubMed] [Google Scholar]
  • 147.Li N, Zhang J, Liang Y, Shao J, Peng F, Sun M, et al. A controversial tumor marker: is SM22 a proper biomarker for gastric cancer cells? J Proteome Res. 2007;6:3304–12. doi: 10.1021/pr0702363. [DOI] [PubMed] [Google Scholar]
  • 148.Tänzer M, Liebl M, Quante M. Molecular biomarkers in esophageal, gastric, and colorectal adenocarcinoma. Aliment Pharmacol Ther. 2013;140:133–47. doi: 10.1016/j.pharmthera.2013.06.005. [DOI] [PubMed] [Google Scholar]
  • 149.de Wit M, Fijneman RJA, Verheul HMW, Meijer GA, Jimenez CR. Proteomics in colorectal cancer translational research: Biomarker discovery for clinical applications. Clin Biochem. 2013;46:466–79. doi: 10.1016/j.clinbiochem.2012.10.039. [DOI] [PubMed] [Google Scholar]
  • 150.Kalantari S, Nafar M, Samavat S, Rezaei-Tavirani M, Rutishauser D, Zubarev R. Urinary Prognostic Biomarkers in Patients With Focal Segmental Glomerulosclerosis. Nephrourol Mon. 2014;6:1–4. doi: 10.5812/numonthly.16806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Pooladi M, Rezaei-Tavirani M, Hashemi M, Hesami-Tackallou S, Khaghani-Razi-Abad S, Moradi A, et al. Cluster and Principal Component Analysis of Human GlioblastomaMultiforme (GBM) Tumor Proteome. Iran J Cancer Prev. 2014;7:87–95. [PMC free article] [PubMed] [Google Scholar]
  • 152.Kalantari S, Nafar M, Rutishauser D, Samavat S, Rezaei-Tavirani M, Yang H, et al. Predictive urinary biomarkers for steroid-resistant and steroid-sensitive focal segmental glomerulosclerosis using high resolution mass spectrometry and multivariate statistical analysis. BMC Nephrology. 2014;15 doi: 10.1186/1471-2369-15-141. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Gastroenterology and Hepatology From Bed to Bench are provided here courtesy of Shahid Beheshti University of Medical Sciences

RESOURCES