Topic Highlight Open Access
Copyright ©2014 Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Oct 14, 2014; 20(38): 13791-13803
Published online Oct 14, 2014. doi: 10.3748/wjg.v20.i38.13791
Potential prognostic, diagnostic and therapeutic markers for human gastric cancer
Ming-Ming Tsai, Department of Nursing, Chang-Gung University of Science and Technology, Taoyuan 333, Taiwan
Chia-Siu Wang, Department of General Surgery, Chang Gung Memorial Hospital at Chiayi, Taoyuan 613, Taiwan
Chung-Ying Tsai, Hsiang-Cheng Chi, Yi-Hsin Tseng, Kwang-Huei Lin, Department of Biochemistry, College of Medicine, Chang-Gung University, Taoyuan 333, Taiwan
Author contributions: Tsai MM, Tsai CY, Chi HC and Tseng YH have substantial contributions to the conception and design, and acquisition of data; Tsai MM wrote and prepared the manuscript; Tsai MM, Wang CS and Lin KH revised the manuscript critically for important intellectual content; Lin KH approved the final version to be published.
Supported by Grants from Chang Gung Memorial Hospital, Taoyuan, Taiwan, No. CMRPF1C0151 and No. CMRPG6D0011
Correspondence to: Kwang-Huei Lin, PhD, Professor, Department of Biochemistry, College of Medicine, Chang-Gung University, 259 Wen-Hwa 1st Road, Kweisan, Taoyuan 333, Taiwan. khlin@mail.cgu.edu.tw
Telephone: +886-3-2118263 Fax: +886-3-2118263
Received: February 28, 2014
Revised: April 18, 2014
Accepted: May 26, 2014
Published online: October 14, 2014

Abstract

The high incidence of gastric cancer (GC) and its consequent mortality rate severely threaten human health. GC is frequently not diagnosed until a relatively advanced stage. Surgery is the only potentially curative treatment. Thus, early screening and diagnosis are critical for improving prognoses in patients with GC. Gastroscopy with biopsy is an appropriate method capable of aiding the diagnosis of specific early GC tumor types; however, the stress caused by this method together with it being excessively expensive makes it difficult to use it as a routine method for screening for GC on a population basis. The currently used tumor marker assays for detecting GC are simple and rapid, but their use is limited by their low sensitivity and specificity. In recent years, several markers have been identified and tested for their clinical relevance in the management of GC. Here, we review the serum-based tumor markers for GC and their clinical significance, focusing on discoveries from microarray/proteomics research. We also review tissue-based GC tumor markers and their clinical application, focusing on discoveries from immunohistochemical research. This review provides a brief description of various tumor markers for the purposes of diagnosis, prognosis and therapeutics, and we include markers already in clinical practice and various forthcoming biomarkers.

Key Words: Tumor marker, Prognosis, Gastric cancer, Serum-based, Tissue-based

Core tip: Serum-based gastric cancer tumor markers and their clinical significance or application are discussed. Serum-based carcinoembryonic antigen, carbohydrate antigen 19-9 and carbohydrate antigen 72-4 and tissue-based human epidermal growth factor receptor 2/Neu are potential tumor markers for various types of gastrointestinal cancer. This review provides a brief description of various tumor markers for the purposes of diagnosis, prognosis and therapeutics, and we include markers already in clinical practice and various forthcoming biomarkers. Hopefully, based on the markers, we will generate accurate diagnoses, prognoses and select the most appropriate therapy.



INTRODUCTION

Gastric cancer (GC) is the second most common cancer worldwide and the sixth leading cause of tumor-associated death in Taiwan. However, the pathogenic mechanisms underlying GC tumorigenesis remain unknown[1]. Surgery continues to be the only curative treatment for GC. In a recent study, more than 30% of surgical patients who presented with GC were too advanced for curative resections. To improve the low survival outcome and assist in the earlier diagnosis of specific tumor types, new prognostic and/or therapeutic tumor markers are required[2]. Indeed, the identification of novel tumor markers presents a new approach for the early screening of populations and for GC therapy.

In this review, we focus primarily on gastric adenocarcinomas. Based on Lauren’s classification, “intestinal-differentiated” (I-GC) and “diffuse-undifferentiated” (D-GC) are two histomorphologic GC subtypes[3,4]. Although these subtypes show related histomorphologic lesions, the tumor cells may contrast in their aggressiveness or response to chemotherapy[5]. The molecular actions involved in the progression of GC are multipart and involve multiple genes and steps that operate sequentially or in concert[4]. Several risk factors for GC, including Helicobacter pylori (H. pylori) infection, genetic alterations, and chromosomal instability, have been reported[6-8]. The identification of various tumor markers has also added to our basic knowledge of molecular and cellular mechanisms of GC tumorigenesis and progression[8]. The majority of tumor markers are effective prognostic tools that are used to identify groups of patients at risk of relapse or metastasis or to monitor cancer survivors following treatment[9]. However, suitable tumor markers to expose the molecular mechanisms of GC or monitor disease development are critical.

Cancer may increase tumor markers in blood or body tissues. There are numerous and varying tumor markers for specific disease processes, which are used to aid in the diagnosis of cancer. An increase in a particular biomarker may indicate cancer. However, there may also be other underlying causes. Tumor markers may be produced directly by cancer cells or by non-cancer cells responding to the tumor. In summary, the diagnostic assay should include gastroscopy with biopsy. Thus, tumor markers are not optimal for screening for GC on a population basis, and as diagnostic assays, they show poor sensitivity and specificity. Assays based on multiple tumor markers will provide more exact results[10]. Thus far, assays based on multiple tumor markers are helpful only as prognostic indicators in patients with GC to monitor cancer survivors following treatment[11-15]. Recently, many studies have been performed using microarrays/proteomics, and they have reported new markers with potential clinical significance in patients with GC. In this paper, we review the progress in serum-based tumor markers associated with GC, focusing on discoveries from microarray/proteomics research[16-20]. The most widely investigated serum-based tumor markers for GC are listed in Table 1.

Table 1 Serum-based tumor markers in gastric cancer.
Biological categoryBiomarker (Gene name)Cutoff level /Expression levels in human GC serumClinical significanceAssociated tumor typesRef.
Oncofetal antigenCEA5 μg/LPrognosisGastrointestinal cancer[24,25]
GC progression monitoringCervix cancer
Lung cancer
Ovary cancer
Breast cancer
Urinary tract cancer
Glycoprotein antigenCA19.937 kU/LPrognosisMainly pancreatic cancer[26]
GC progressionbut also colorectal cancer
monitoringother types of gastrointestinal cancer
CA72.46 kU/LPrognosisOvary cancer[27]
GC progressionBreast cancer
monitoringColon cancer
Pancreatic cancer
CytokeratinsCyfra21.1VariablePrognosisMany types of carcinoma[29]
Some types of sarcoma
TPAVariablePrognosisCarcinoma[30]
TPSVariablePrognosisAdult epithelial tumours[30]
HormoneΒ subunit of hCG4 μg/LPrognosisSeminoma[31]
Choriocarcinoma
Germ cell tumors
Choriocarcinoma
Islet cell tumor
Testicular cancer
Cell proliferationReg IVOverexpressionPrognosisGastrointestinal malignancies[33,34]
GranulinOverexpressionPrognosisGastrointestinal malignancies[35,36]
Signaling protein14-3-3 proteinDownexpressionPrognosisEpithelial cancer[40-42]
Lung cancer
Breast cancer
Bladder cancer
Liver cancer
Oral cancer
Head-and-neck squamous cell cancer
DKK-1OverexpressionPrognosisProstate cancer[43-49]
Breast cancer
Colon cancer
Esophageal cancer
Lung cancer
Multiple myeloma (MM)
InflammatorySAAOverexpressionPrognosisLung cancer[50,52,53]
Kidney cancer
Colon cancer
Prostate cancer
Liver cancer
Cell apoptosisMG7-AgOverexpressionPrognosisGastrointestinal cancer[55,56]
EnzymePGOverexpressionPrognosisGastrointestinal cancer[57]
CARCINOEMBRYONIC ANTIGEN

Carcinoembryonic antigen (CEA) is a cell-surface-anchored protein involved in cell-cell adhesions. CEA serves as a functional receptor for colon cancer E-selectin and L-selectin ligands, which may be critical for the metastatic spreading of colon cancer cells[21,22]. Serum from individuals with colon cancer often shows higher levels of CEA than that from healthy controls. CEA from serum is primarily used as a biomarker to monitor colon cancer treatment, to identify recurrences following surgical resection and to stage or localize cancer spread[23].

CEA levels may also be raised in other cancer types and in some non-neoplastic conditions. Several factors may influence increases in CEA. It is associated with tumor grade, lymph node metastasis, distant metastasis and tumor stage, suggesting that as the tumor progresses, CEA levels increase[24].

Anti-CEA antibodies are also frequently used in IHC to identify cells expressing CEA in tissue samples. In adults, CEA is expressed only in cancer cells, primarily adenocarcinomas. It can therefore be used to distinguish between this and other similar types of cancers. Because even anti-CEA antibodies tend to show some degree of cross-reactivity, false positive results are observed, and this assay is typically used in combination with other tests[25].

CARBOHYDRATE ANTIGEN 19-9

In 1981, carbohydrate antigen 19-9 (CA19-9) (also called the Lewis antigen) was first discovered in the serum of individuals with colon or pancreatic cancer. However, CA19-9 is used primarily in the management of pancreatic cancer. CA19-9 may be increased in many types of gastrointestinal cancer[26].

CARBOHYDRATE ANTIGEN 72-4

Carbohydrate antigen 72.4 (CA72.4) is a mucin-like glycoprotein found on the surface of many cancer cells[27]. CA72.4-Ab assay shows good specificity for GC and is used to identify GC relapses and for follow-up after treatment[28].

CYTOKERATIN SUBUNIT 19 FRAGMENT, TISSUE POLYPEPTIDE ANTIGENAND TISSUE POLYPEPTIDE-SPECIFIC ANTIGEN

Cytokeratin subunit 19 fragment (Cyfra21.1) is potentially useful for monitoring lung carcinoma. It is a member of the keratin family. The keratins are intermediate filament proteins responsible for the structural integrity of epithelial cells and are grouped into cytokeratins and hair keratins. Cyfra21.1 has been suggested to play a role in lung, colon, stomach, pancreas, breast, and prostate cancers. Cyfra21.1 is the most sensitive tumor marker for non-squamous cell lung cancer. Because Cyfra21.1 detects only fragments of cytokeratin 19, the test shows higher specificity than tissue polypeptide antigen (TPA) and is an independent prognostic factor[29].

The TPA tumor marker comprises a molecular complex of cytokeratins 8, 18, and 19. It is used in the diagnosis and staging of bronchogenic cancer. TPA assays represent first-generation cytokeratin tumor marker tests.

Although tissue polypeptide-specific antigen has been reported to be a potentially useful serum marker in adult epithelial tumors, few reports have been published on childhood malignancies. Currently, there is no widely used marker for Wilms’ tumor[30].

β-SUBUNIT OF HUMAN CHORIONIC GONADOTROPIN

β-subunit of human chorionic gonadotropin (β-hCG) is a hormone formed by the syncytiotrophoblast, a component of the fertilized egg, following conception. Following grafting, the syncytiotrophoblast gives rise to the placenta. Some tumors produce β-hCG; thus, elevated levels in individuals who are not pregnant may indicate cancer. However, whether β-hCG production promotes carcinogenesis remains unknown.

β-hCG may be used as a tumor marker as its β subunit is secreted by some tumors. Based on this reason, a positive result in males indicates testicular cancer. Combined with α-fetoprotein, β-hCG is a good tumor marker for monitoring of tumors of germ cells[31].

REGENERATING GENE IV

Regenerating gene IV (Reg IV) belongs to the regenerating family of secreted C-lectin proteins[32]. Reg proteins are expressed normally in the gastrointestinal tract and are induced in inflammatory bowel disease and in some gastrointestinal malignancies. They show multiple functions including enhancing tissue regeneration, proliferation and are anti-apoptotic[33]. Reg IV is expressed at low levels in a subgroup of primary tumors and is moderately or highly expressed in a majority of hormone refractory and metastatic tumors[34].

GRANULIN

Granulins are a family of secreted, glycosylated peptides that are cleaved from a single precursor. Both the peptides and the intact granulins regulate cell growth. However, different members of the granulin family may act as inhibitors, activators, or show dual roles in cell growth. The granulin family is important for normal development, wound healing, and in tumorigenesis[35]. In addition, the human liver contributes to the development of liver cancer by secreting a granulin-like growth hormone[36].

14-3-3 PROTEINS

14-3-3 proteins, which belong to a family of regulatory proteins, are able to bind a multitude of signaling proteins of various functions, such as kinases, phosphatases, and transmembrane receptors. More than 200 signaling proteins have been reported as being ligands for 14-3-3 proteins[37,38].

Alterations in the expression of several 14-3-3 proteins have been associated with some human cancers. The down-regulation of 14-3-3r has been reported in a multitude of human epithelial cancers. Recently, Urano et al[39] showed that breast cancer cells reduced 14-3-3r protein levels by upregulating the protein Efp, a ring-finger-dependent ubiquitin ligase (E3), which targets 14-3-3r for ubiquitin-mediated degradation by the proteosome. MCF7 cells treated with antisense Efp constructs showed increased levels of 14-3-3r, and athymic mice transplanted with these cells showed decreased tumors. Similar trends of cancer-associated downregulation of 14-3-3r have been reported in human lung cancer, vulva squamous neoplasias, bladder cancer, liver cancer, oral cancer, and head-and-neck squamous cell cancer, suggesting a common role for 14-3-3r as a tumor suppressor[40-42].

DICKKOPF-1

Dickkopf-1 (DKK1) inhibits Wnt/β-catenin signaling, which is essential for embryonic head development. DKK1 regulates Wnt signaling by binding to the Wnt coreceptor lipoprotein-related protein-5 (LRP5). Overexpression or down-regulation of DKK1 gene expression levels has been observed at various stages of tumorigenesis in multiple cancers, such as breast, colorectal, prostate, esophageal, lung cancers, and multiple myeloma (MM). Thus, DKK1 play dual roles as an oncogene or tumor suppressor. Serum from individuals with breast cancer and bone metastases showed significantly higher DKK1 levels compared with healthy individuals. In addition, human primary lung and esophageal cancers, lung cancer cell lines, and esophageal cancer cell lines show increased DKK1 expression. However, DKK1 protein expression was also down-regulated in colon cancer and melanoma[43-49]. Qian et al[44] reported that DKK1 protein expression was detected in the majority of MM cell lines and clinical MM samples.

SERUM AMYLOID A PROTEIN

Serum amyloid A (SAA) proteins combined with the high-density lipoprotein (HDL) complex belong to a family of apolipoproteins that are secreted during the acute phase of inflammation[50]. There are three isoforms of SAA. SAA (SAA1 and SAA2) levels are consistently increased in the liver during acute inflammation, and SAA3 is induced in various distinct tissues[50,51].

SAA are not specific for any type of cancer and are expected to be elevated in other malignant diseases and in inflammatory diseases[52,53]. Cho et al observed that SAA had previously been reported to increase in several different cancers including kidney, colon, prostate cancers, leukemias and lymphomas. However, these proteins are released into the blood from the liver. These findings suggest that these proteins may not be of much clinical significance in our future efforts to more effectively diagnose and monitor cancer[50].

GASTRIC CANCER ASSOCIATED ANTIGEN (MG7-AG)

Akashi et al[54] produced a MG7-Ag monoclonal antibody to use for GC screening. Using the immuno-polymerase chain reaction (Immuno-PCR) assay, the authors observed that MG7-Ag had the highest sensitivity and specificity for detecting GC. They tested serum MG7-Ag levels and found that the rate of MG7-Ag positivity was 82.8% for GC in 198 GC patients and 44.4% for colonic cancer. However, the Immuno-PCR assay is too complicated and expensive for screening the high-risk population in Linqu county. Therefore, they used an ELISA to detect MG7-Ag expression in serum from patients with GC or other carcinomas. They also used IHC to test MG7-Ag expression in GC tissues[55,56].

PEPSINOGENS

There are three genes that encode human pepsinogen A. A fourth human gene encodes gastricsin, also known as pepsinogen C, a digestive enzyme. Gastricsin is an aspartic proteinase that belongs to the A1 peptidase family and is produced in the stomach; it constitutes a major component of the gastric mucosa. Gastricsin is secreted into the serum and is synthesized as an inactive zymogen that includes a highly basic prosegment. Gastricsin is converted into its active mature form at low pH by serial cleavage of the prosegment, performed by the enzyme. Polymorphisms in gastricsin are associated with susceptibility to GC. Serum levels of the enzyme have been identified as tumor markers for specific GCs and H. pylori-associated gastritis[57].

Tumor markers may also be detected using IHC. Although IHC remains the most reliable and cheapest method, several efforts have been made to identify and test novel tumor markers to achieve the prognosis goals. In recent years, many proteins have been identified and validated for their clinical significance in the management of GC. The most widely investigated tissue-based tumor markers for GC and the most correlated clinical parameters are listed in Table 2.

Table 2 Tissue-based tumor markers in gastric cancer.
Biological categoryBiomarker (Gene name)ParametersClinical significanceAssociated tumor typesRef.
ReceptorHER2/neuLauren histotypePrognosisOvarian cancer[58-63]
Lymph node metastasisTherapeutic responseStomach cancer
Uterine cancer
Endometrial cancer
Breast cancer
Esophageal cancer
VEGFLauren histotypePrognosisBreast cancer.[64-68]
Tumor progressionTherapeutic responseColon cancer
Gastric cancer
Liver cancer
Lung cancer
Ion channelhERG1Lauren histotypePrognosisEndometrial cancer[69]
GradingGastric cancer
StageColorectal cancer
Transcription factorKLF5/IKLF/BTEB2GradingPrognosisBladder cancer[71,72]
StageProstate cancer
Lymph node statusBreast cancer
Colon cancer
SATB1Lymph node metastasisPrognosisBreast cancer[75-78]
Distant metastasisGastric cancer
StageColon cancer
Bladder cancer
c-myc2Lymph node metastasisPrognosisCervix cancer[80]
Colon cancer
Breast cancer
Lung cancer
Stomach cancer
EnzymeCA IXLymph node metastasisPrognosisKidney cancer[81-83]
Colon cancer
Cervical cancer
MMP-2StagePrognosisOvarian cancer[79,80,84,85]
SurvivalColon cancer
Breast cancer
Bladder cancer
Gastric cancer
HDACLymph node metastasisPrognosisRenal cancer[86-92]
SurvivalColorectal cancer
Gastric cancer
Breast cancer
Pancreatic cancer
Liver cancer
Lung cancer
Prostate cancer
Hodgkin's lymphoma
COX-2Lymph node metastasisPrognosisBreast cancer[93,94]
SurvivalIntestinal tract cancer
SLPIDepth of invasionPrognosisOvan cancer[96,97]
Serosal invasionCervical cancer
Lymph node metastasisGastric cancer
Stage
GLO1Depth of invasionPrognosisColon cancer[99-103]
Serosal invasionBreast cancer
Lymph node metastasisProstate cancer
StageGastric cancer
Melanoma cancer
Cell growthKi-67Lymph node metastasisPrognosisProstate cancer[104]
Brain cancer
Breast cancer
Nephroblastoma
TGF-βStagePrognosisBreast cancer[105,106]
Gastric cancer
SignalPKP3StagePrognosisGastric cancer[109,110]
Protein/cell adhesionLung cancer
E-cadherinInvasionPrognosisEsophagus cancer[111]
GradingOvary cancer
Lauren histotypeStomach cancer
TSP-1StagePrognosisBreast cancer[112-115]
Melanoma cancer
Lung cancer
Gastric cancer
SPARCDepth of invasionPrognosisBreast cancer[118-120]
Serosal invasionMelanoma cancer
Lymph node metastasisGastric cancer
Distant metastasisOesophageal cancer
Glioma cancer
Regulate cell death (apoptosis)Bcl-2Lymph node metastasisPrognosisMelanoma cancer[121]
Breast cancer
Prostate cancer
Chronic lymphocytic Leukemia lung cancer
Bcl-6Lymph node metastasisPrognosisGastric cancer[122]
Pancreas cancer
Colon cancer
Bladder cancer
BaxNo dataPrognosisGastric cancer[123-125]
Colon cancer
CytokineCXCL1/CXCR2Lymph node metastasisPrognosisGastric cancer[128]
Colon cancer
Vesicular transportARF1Depth of invasionPrognosisGastric cancer[129]
Serosal invasion
Lymph node metastasis
Distant metastasis
Stage
HUMAN EPIDERMAL GROWTH FACTOR RECEPTOR 2/NEU

Human epidermal growth factor receptor 2/neu (HER2/neu) is a well-known oncogene. HER2/neu belongs to a member of the epidermal growth factor receptor family and plasma membrane-bound receptor tyrosine kinases. Overexpression of this oncogene has been shown to play an important role in the tumorigenesis of aggressive types of breast cancer. Thus, the protein has become an important tumor marker and target of therapy for over 30% of patients with breast cancer. It is positively associated with increased breast cancer recurrence and poor prognosis. Overexpression of HER2/neu has also been reported in ovarian, stomach, uterine and endometrial cancers. Dimerization results in the autophosphorylation of tyrosine residues within the cytoplasmic domain of the receptors and initiates a variety of signaling pathways.

IHC is used to measure levels of HER2/neu protein in samples. Fluorescence in situ hybridization (FISH) assays may also be used to measure whether HER2/neu gene amplification has occurred. The extracellular domain of HER2 may be shed from the surface of tumor cells and enter the circulation. Testing serum for HER2 using enzyme-linked immunosorbent assay (ELISA) offers a method for determining HER2 levels. Changes in serum HER2 concentrations may be useful in predicting response to trastazumab treatment[58-63].

VASCULAR ENDOTHELIAL GROWTH FACTOR

Vascular endothelial growth factor (VEGF) is an angiogenic factor. It is a signaling protein secreted by many solid tumors. VEGF overexpression has been reported in many human cancer cells including breast, colon, gastric, liver and lung cancers. It is also expressed in stromal cells, particularly at sites of relative hypoxia. VEGF enhances endothelial cell proliferation and migration and induces endothelial cell angiogenesis. VEGF is also called vascular permeability factor. Thus, VEGF increases vascular permeability to plasma and its proteins, a typical requirement of tumor microvasculature. IHC and ELISA are popular methods used to examine expression patterns of VEGF in serum and tissues from cancer patients[64-68].

HUMAN ETHER-À-GO-GO-RELATED GENE

Human ether-à-go-go-related gene (hERG1) belongs to the K+ channel family, which may regulate cell apoptosis and proliferation. Overexpression of this protein has been observed in several tumors, such as primary human endometrial, gastric and colorectal cancers. hERG1 gene expression has been used as a specific tumor marker in colon cancer, and it regulates the invasion of tumor cells[69].

KRUEPPEL-LIKE FACTOR 5 (KLF5/IKLF/BTEB2)

KLF5/IKLF/BTEB2 is a member of the Kruppel-like transcription factor family. Several members of the KLF family have been implicated in the development of human cancers[70]. KLF5 repeatedly shows genetic deletions and decreased expression in human prostate and breast cancers[71,72]. KLF5 also plays a suppressive role in tumor growth in colon cancer. However, overexpression of KLF5 activated fibroblast cell growth and tumorigenesis. KLF5 has also been reported to affect H-Ras, Wnt-1, platelet-derived growth factor A chain (PDGF), cyclin D1, PPARg and ERBB2 oncogenes[73]. Moreover, several growth factors, such as PMA, sphingosine-1-phosphate, and β-FGF, induce the expression of KLF5. Furthermore, KLF5 has been reported to promote angiogenesis in a knock-out mouse model. In a bladder cancer cell line, KLF5 was reported to inhibit cell growth. These findings suggest that the actual role of KLF5 in tumorigenesis remains unknown[74].

SPECIAL AT-RICH SEQUENCE-BINDING PROTEIN-1

Special at-rich sequence-binding protein-1 (SATB1) is an AT-rich sequence-binding protein 1 and gene regulator. It functions as a genome organizer. SATB1 is highly expressed in many types of human cancers including breast, gastric, colon and bladder carcinomas. It enables tumor growth and metastasis by altering the expression of a large number of genes[75-78].

C-MYC2

c-Myc protein is a transcription factor that activates and represses the expression of many genes. In addition, c-myc has three isoforms and is extremely influential in controlling DNA replication[79].

The MAPK/ERK pathway may activate c-myc, and c-myc activation results in many cellular effects including cell proliferation, apoptosis, differentiation and stem cell self-renewal. c-Myc is a very robust proto-oncogene, and it is very often upregulated in many cancers. c-Myc is correlated with recurrence and may be a potential prognostic factor. High levels of c-myc2 were correlated with lymph node metastasis and poor prognosis in cancers of the cervix, colon, breast, lung and stomach[80].

CARBONIC ANHYDRASE 9

Carbonic anhydrases (CAs) are a large family of zinc metalloenzymes that catalyze the reversible hydration of carbon dioxide. They are involved in some biological reactions, such as respiration, calcification, acid-base balance, bone resorption, and the formation of aqueous humor, cerebrospinal fluid (CSF), saliva, and gastric acid. CA9 is a transmembrane protein and a tumor-associated carbonic anhydrase isoenzyme. It is increased in renal cancers and hypoxic solid tumors. It is also involved in cell proliferation and tumorigenesis.

Drug compounds targeted to CA9 are in preclinical development for the treatment of kidney, colon, and cervical cancers. CA9 is one of the markers of tumor hypoxia. Moreover, recent studies assaying CA9 levels and various clinicopathological outcomes indicated that CA9 expression may also be a valuable prognostic factor of overall survival. Antibodies targeted against CA9 may be used to show hypoxic regions in many solid tumors[81-83].

MATRIX METALLOPROTEINASE-2

Matrix metalloproteinase-2 (MMP-2) is a collagenase that assists tumor growth and invasion by digesting the extracellular matrix surrounding the tissue. MMP-2 is considered a possible tumor marker for ovarian, colon, breast, bladder and gastric cancers[79,80,84,85].

HISTONE DEACETYLASES

Histone deacetylases (HDACs) promote the removal of acetyl groups from acetylated residues to release an acetate molecule. Lysine acetylation typically occurs in a number of non-histone proteins.

Mutation and/or aberrant expression of various HDACs have often been observed in cancer, making them important therapeutic targets for many human cancers. Several studies have indicated that overexpression of HDACs is present in renal, colorectal, gastric, breast, pancreatic, liver, lung and prostate cancers and in classical Hodgkin’s lymphoma and that they are associated with poor prognoses[86-92].

CYCLOOXYGENASE-2

Cyclooxygenase (COX)-2 is an enzyme responsible for inflammation and pain. COX-2 appears to be associated with cancers and irregular growths in the intestinal tract[93]. COX inhibitors have been shown to reduce the occurrence of cancers. COX-2 inhibitors are currently being studied for the treatment of breast cancer[94].

ANTIPROTEASE SECRETORY LEUKOCYTE PROTEASE INHIBITOR

Secretory leukocyte protease inhibitor (SLPI) neutralizes elastase, tryptase, and cathepsin G[95]. Several studies have indicated that SLPI may be involved in tumorigenesis[96]. Abundant expression of SLPI was noted in several cancers including ovarian, cervical and gastric cancers[97].

GLYOXALASE I

Glyoxalase I (GLO1) is an essential component in pathways leading to the detoxification of methylglyoxal (MG). Over-expression of GLO1 has been reported in several cancers, such as colon, breast, prostate, gastric cancers and melanoma[98-103].

KI-67

Ki-67 is a nuclear protein that is a cellular marker of cell proliferation. It always correlates with cell proliferation. During interphase, Ki-67 may be exclusively detected within the cell nucleus, and when the cell enters mitosis, the majority of Ki-67 relocates to the surface of the chromosomes. Ki-67 is present during all active phases of the cell cycle (G1, S, G2, and mitosis), but is absent in resting cells (G0). Thus, Ki-67 is a good marker for determining cell growth rate. Ki-67-positive tumor cells are often correlated with advanced tumors including carcinomas of the prostate, brain, breast and nephroblastomas. With respect to these tumors, survival and tumor recurrence have been positively correlated with Ki-67 IHC staining[104].

TRANSFORMING GROWTH FACTOR-β

Transforming growth factor-β (TGF-β) regulates cellular proliferation, differentiation and apoptosis. TGF-β is a multifunctional cytokine, and increased or decreased production has been linked to many cancers including breast cancer and GC[105].

TGF-β is a pluripotent cytokine with diverse effects on normal physiology and a role in both normal mammary gland development and the progression of breast cancer. During early stages of cancer, TGF-β acts as a tumor suppressor, whereas in later stages, when tumor cells become resistant to growth inhibition by TGF-β, it acts as a tumor promoter. Thus, TGF-β as a tumor marker is useful primarily during the follow-up of cancer patients and particularly in monitoring advanced disease[106].

PLAKOPHILIN-3

Plakophilin-3 (PKP3) is a member of the armadillo (ARM)/plakophilin families. ARM-related proteins physically localize to cell desmosomes and nuclei and are involved in linking cadherins to intermediate filaments in the cytoskeleton. PKP3 may play a role as in cellular desmosome-dependent adhesion and signaling pathways[107,108]. Several publications have reported that genetic aberrations in members of the ARM-protein family including plakoglobin (PKGB), h-catenin (CTNNB1), and adenomatous polyposis coli (APC) promote tumor progression. However, with regard to the role(s) of the subfamily member PKP3 during carcinogenesis, evidence shows that PKP3 is a potential prognostic marker and that it shows clinicopathological correlation with lung cancer and GC[109,110].

E-CADHERIN

E-cadherin is a calcium-regulated transmembrane cell-cell adhesion protein that is expressed in most normal epithelial tissues. Selective loss of E-cadherin can cause dedifferentiation and invasiveness in human carcinomas, suggesting that E-cadherin may play a role as a tumor suppressor. Reduced expression of E-cadherin has been observed in aggressive tumors of the esophagus, ovary, and stomach[111].

THROMBOSPONDIN 1

Thrombospondin 1 (TSP-1) is an extracellular matrix glycoprotein that regulates cell adhesion, cell motility, angiogenesis, and cell growth. TSP1 is down-regulated in many tumors including breast, melanoma, lung and gastric cancers[112-115].

SECRETED PROTEIN AND RICH IN CYSTEINE

Secreted protein and rich in cysteine (SPARC) is a member of the matricellular protein family. SPARC may regulate cell-matrix interactions and cell function without participating in the structural scaffold of the extracellular matrix[116,117]. Recently, SPARC over-expression has been reported in breast, melanoma, gastric, esophageal cancers and gliomas[118-120].

B-CELL LYMPHOMA 2/B-CELL LYMPHOMA 6/BCL-2-ASSOCIATED X PROTEIN

B-cell lymphoma 2 (Bcl-2) is a member of the Bcl-2 family of regulator proteins that either induce or inhibit cell apoptosis. Bcl-2 is an important anti-apoptotic oncoprotein. Mutation of the Bcl-2 gene causes cancers including melanoma, chronic lymphocytic leukemia, breast, gastric, prostate, and lung cancers[121]. Bcl-2 is a commonly used IHC marker, and it has been used in the classification of lymphoid processes and the identification of specific epithelial tumors.

Bcl-6 is also a biomarker of gastric, pancreas, colon and bladder cancers[122].

Bcl-2-associated X protein (BAX) is a member of the Bcl-2 gene family. BAX is a pro-apoptotic factor based on its binding to Bcl-2. It is also a prognostic marker of colon and bladder cancers[123-125].

CXCL1/CXCR2

CXCL1 is thought to be a chemo-attractant for neutrophils and lymphocytes[126,127]. However, it was observed that neutrophil infiltration into tumors was dependent on CXCR2 signaling. Moreover, the expression of CXCL1/CXCR2 was significantly associated with GC progression. This evidence is consistent with the previous observation the CXCL1/CXCR2 signaling pathway was important in the metastasis of several cancers including breast, gastric and colon cancers[128].

ADP-RIBOSYLATION FACTOR 1

ADP-ribosylation factor 1 (ARF1) is a member of the small GTPase family. ARF1 is involved in many cellular processes including cell differentiation, proliferation, necrosis, apoptosis, and inter- and intracellular signaling[129].

CONCLUSION

There are no current excellent tumor indices or tumor markers for GC. However, it is very important to use many tumor markers in different types of cancer to screen, diagnose and stage the cancer, for evaluation of prognosis and for monitoring of recurrence following treatment. However, determining the significance of the markers and assessing and interpreting them requires an accumulation of expertise and experience. These processes cannot work when based on a single tumor marker. From the viewpoint of the clinic, it is critical to examine the performance of the marker using imaging, to select a tumor marker assay and to consider the variety of non-specific factors that may have caused the increase in the biomarker. Finally, this knowledge may lead to more accurate diagnoses and prognoses, may enable the selection of optimal and appropriate therapies and also may enable assessments of the effectiveness of treatments, with or without postoperative tracking of recurrence. Thus, tumor markers are only references; they are used by the physician for examining the cancer and cannot diagnose the disease themselves.

Footnotes

P- Reviewer: Kariya S, Kim H S- Editor: Nan J L- Editor: Wang TQ E- Editor: Ma S

References
1.  Wang CS, Hsieh CC, Chao TC, Jan YY, Jeng LB, Hwang TL, Chen MF, Chen PC, Chen JS, Hsueh S. Resectable gastric cancer: operative mortality and survival analysis. Chang Gung Med J. 2002;25:216-227.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Marrelli D, Roviello F, De Stefano A, Farnetani M, Garosi L, Messano A, Pinto E. Prognostic significance of CEA, CA 19-9 and CA 72-4 preoperative serum levels in gastric carcinoma. Oncology. 1999;57:55-62.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Laurén P. Histogenesis of intestinal and diffuse types of gastric carcinoma. Scand J Gastroenterol Suppl. 1991;180:160-164.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
4.  Tahara E. Molecular mechanism of stomach carcinogenesis. J Cancer Res Clin Oncol. 1993;119:265-272.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 228]  [Cited by in F6Publishing: 248]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
5.  Cervantes A, Rodríguez Braun E, Pérez Fidalgo A, Chirivella González I. Molecular biology of gastric cancer. Clin Transl Oncol. 2007;9:208-215.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 41]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
6.  Barreto-Zuñiga R, Maruyama M, Kato Y, Aizu K, Ohta H, Takekoshi T, Bernal SF. Significance of Helicobacter pylori infection as a risk factor in gastric cancer: serological and histological studies. J Gastroenterol. 1997;32:289-294.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 38]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
7.  Kim KM, Kwon MS, Hong SJ, Min KO, Seo EJ, Lee KY, Choi SW, Rhyu MG. Genetic classification of intestinal-type and diffuse-type gastric cancers based on chromosomal loss and microsatellite instability. Virchows Arch. 2003;443:491-500.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 52]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
8.  Yasui W, Oue N, Kuniyasu H, Ito R, Tahara E, Yokozaki H. Molecular diagnosis of gastric cancer: present and future. Gastric Cancer. 2001;4:113-121.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 82]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
9.  Allgayer H, Heiss MM, Schildberg FW. Prognostic factors in gastric cancer. Br J Surg. 1997;84:1651-1664.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 58]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
10.  Pesch B, Taeger D, Johnen G, Gawrych K, Bonberg N, Schwentner C, Wellhäußer H, Kluckert M, Leng G, Nasterlack M. Screening for bladder cancer with urinary tumor markers in chemical workers with exposure to aromatic amines. Int Arch Occup Environ Health. 2014;87:715-724.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 25]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
11.  Dilege E, Mihmanli M, Demir U, Ozer K, Bostanci O, Kaya C, Aksakal O, Sakiz D. Prognostic value of preoperative CEA and CA 19-9 levels in resectable gastric cancer. Hepatogastroenterology. 2010;57:674-677.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 106]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
12.  Duraker N, Celik AN. The prognostic significance of preoperative serum CA 19-9 in patients with resectable gastric carcinoma: comparison with CEA. J Surg Oncol. 2001;76:266-271.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 62]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
13.  Emoto S, Ishigami H, Yamashita H, Yamaguchi H, Kaisaki S, Kitayama J. Clinical significance of CA125 and CA72-4 in gastric cancer with peritoneal dissemination. Gastric Cancer. 2012;15:154-161.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 107]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
14.  Li Y, Yang Y, Lu M, Shen L. Predictive value of serum CEA, CA19-9 and CA72.4 in early diagnosis of recurrence after radical resection of gastric cancer. Hepatogastroenterology. 2011;58:2166-2170.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 32]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
15.  Ychou M, Duffour J, Kramar A, Gourgou S, Grenier J. Clinical significance and prognostic value of CA72-4 compared with CEA and CA19-9 in patients with gastric cancer. Dis Markers. 2000;16:105-110.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 89]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
16.  Lu HB, Zhou JH, Ma YY, Lu HL, Tang YL, Zhang QY, Zhao CH. Five serum proteins identified using SELDI-TOF-MS as potential biomarkers of gastric cancer. Jpn J Clin Oncol. 2010;40:336-342.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 26]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
17.  Liu W, Gao X, Cai Q, Li J, Zhu Z, Li C, Yao X, Yang Q, Xiang M, Yan M. Identification of novel serum biomarkers for gastric cancer by magnetic bead. Front Biosci (Elite Ed). 2010;2:961-971.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
18.  Umemura H, Togawa A, Sogawa K, Satoh M, Mogushi K, Nishimura M, Matsushita K, Tanaka H, Takizawa H, Kodera Y. Identification of a high molecular weight kininogen fragment as a marker for early gastric cancer by serum proteome analysis. J Gastroenterol. 2011;46:577-585.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 28]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
19.  Ahn HS, Shin YS, Park PJ, Kang KN, Kim Y, Lee HJ, Yang HK, Kim CW. Serum biomarker panels for the diagnosis of gastric adenocarcinoma. Br J Cancer. 2012;106:733-739.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 39]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
20.  Zhang Z, Li M, Zhang G, Fang P, Yao H, Xiao Z, Chen Z. Identification of human gastric carcinoma biomarkers by differential protein expression analysis using 18O labeling and nanoLC-MS/MS coupled with laser capture microdissection. Med Oncol. 2010;27:296-303.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 15]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
21.  Thomas SN, Zhu F, Schnaar RL, Alves CS, Konstantopoulos K. Carcinoembryonic antigen and CD44 variant isoforms cooperate to mediate colon carcinoma cell adhesion to E- and L-selectin in shear flow. J Biol Chem. 2008;283:15647-15655.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 135]  [Cited by in F6Publishing: 132]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
22.  Konstantopoulos K, Thomas SN. Cancer cells in transit: the vascular interactions of tumor cells. Annu Rev Biomed Eng. 2009;11:177-202.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 161]  [Cited by in F6Publishing: 167]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
23.  Thomas SN, Tong Z, Stebe KJ, Konstantopoulos K. Identification, characterization and utilization of tumor cell selectin ligands in the design of colon cancer diagnostics. Biorheology. 2009;46:207-225.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 36]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
24.  Maestranzi S, Przemioslo R, Mitchell H, Sherwood RA. The effect of benign and malignant liver disease on the tumour markers CA19-9 and CEA. Ann Clin Biochem. 1998;35:99-103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 94]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
25.  Duffy MJ, van Dalen A, Haglund C, Hansson L, Klapdor R, Lamerz R, Nilsson O, Sturgeon C, Topolcan O. Clinical utility of biochemical markers in colorectal cancer: European Group on Tumour Markers (EGTM) guidelines. Eur J Cancer. 2003;39:718-727.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 257]  [Cited by in F6Publishing: 276]  [Article Influence: 13.1]  [Reference Citation Analysis (0)]
26.  Koprowski H, Herlyn M, Steplewski Z, Sears HF. Specific antigen in serum of patients with colon carcinoma. Science. 1981;212:53-55.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 413]  [Cited by in F6Publishing: 439]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
27.  Louhimo J, Alfthan H, Stenman UH, Haglund C. Serum HCG beta and CA 72-4 are stronger prognostic factors than CEA, CA 19-9 and CA 242 in pancreatic cancer. Oncology. 2004;66:126-131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 64]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
28.  Louhimo J, Kokkola A, Alfthan H, Stenman UH, Haglund C. Preoperative hCGbeta and CA 72-4 are prognostic factors in gastric cancer. Int J Cancer. 2004;111:929-933.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 47]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
29.  Kanazawa H, Yoshikawa T, Yamada M, Shoji S, Fujii T, Kudoh S, Hirata K, Yoshikawa J. CYFRA 21-1, a cytokeratin subunit 19 fragment, in bronchoalveolar lavage fluid from patients with interstitial lung disease. Clin Sci (Lond). 1998;94:531-535.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
30.  D'Alessandro R, Roselli M, Ferroni P, Mariotti S, Spila A, Aloe S, Carone MD, Abbolito MR, Carlini S, Perri P. Serum tissue polypeptide specific antigen (TPS): a complementary tumor marker to CA 15-3 in the management of breast cancer. Breast Cancer Res Treat. 2001;68:9-19.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 26]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
31.  Gregory JJ, Finlay JL. Alpha-fetoprotein and beta-human chorionic gonadotropin: their clinical significance as tumour markers. Drugs. 1999;57:463-467.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 43]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
32.  Hartupee JC, Zhang H, Bonaldo MF, Soares MB, Dieckgraefe BK. Isolation and characterization of a cDNA encoding a novel member of the human regenerating protein family: Reg IV. Biochim Biophys Acta. 2001;1518:287-293.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 128]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
33.  Macadam RC, Sarela AI, Farmery SM, Robinson PA, Markham AF, Guillou PJ. Death from early colorectal cancer is predicted by the presence of transcripts of the REG gene family. Br J Cancer. 2000;83:188-195.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 61]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
34.  Gu Z, Rubin MA, Yang Y, Deprimo SE, Zhao H, Horvath S, Brooks JD, Loda M, Reiter RE. Reg IV: a promising marker of hormone refractory metastatic prostate cancer. Clin Cancer Res. 2005;11:2237-2243.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 40]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
35.  Baker M, Mackenzie IR, Pickering-Brown SM, Gass J, Rademakers R, Lindholm C, Snowden J, Adamson J, Sadovnick AD, Rollinson S. Mutations in progranulin cause tau-negative frontotemporal dementia linked to chromosome 17. Nature. 2006;442:916-919.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1401]  [Cited by in F6Publishing: 1460]  [Article Influence: 81.1]  [Reference Citation Analysis (0)]
36.  Smout MJ, Laha T, Mulvenna J, Sripa B, Suttiprapa S, Jones A, Brindley PJ, Loukas A. A granulin-like growth factor secreted by the carcinogenic liver fluke, Opisthorchis viverrini, promotes proliferation of host cells. PLoS Pathog. 2009;5:e1000611.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 140]  [Cited by in F6Publishing: 139]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
37.  Obsil T, Ghirlando R, Klein DC, Ganguly S, Dyda F. Crystal structure of the 14-3-3zeta: serotonin N-acetyltransferase complex. a role for scaffolding in enzyme regulation. Cell. 2001;105:257-267.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 304]  [Cited by in F6Publishing: 294]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
38.  Cann KL, Hicks GG. Regulation of the cellular DNA double-strand break response. Biochem Cell Biol. 2007;85:663-674.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 73]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
39.  Urano T, Saito T, Tsukui T, Fujita M, Hosoi T, Muramatsu M, Ouchi Y, Inoue S. Efp targets 14-3-3 sigma for proteolysis and promotes breast tumour growth. Nature. 2002;417:871-875.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 282]  [Cited by in F6Publishing: 283]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
40.  Moreira JM, Gromov P, Celis JE. Expression of the tumor suppressor protein 14-3-3 sigma is down-regulated in invasive transitional cell carcinomas of the urinary bladder undergoing epithelial-to-mesenchymal transition. Mol Cell Proteomics. 2004;3:410-419.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 66]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
41.  Iwata N, Yamamoto H, Sasaki S, Itoh F, Suzuki H, Kikuchi T, Kaneto H, Iku S, Ozeki I, Karino Y. Frequent hypermethylation of CpG islands and loss of expression of the 14-3-3 sigma gene in human hepatocellular carcinoma. Oncogene. 2000;19:5298-5302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 183]  [Cited by in F6Publishing: 194]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
42.  Gasco M, Sullivan A, Repellin C, Brooks L, Farrell PJ, Tidy JA, Dunne B, Gusterson B, Evans DJ, Crook T. Coincident inactivation of 14-3-3sigma and p16INK4a is an early event in vulval squamous neoplasia. Oncogene. 2002;21:1876-1881.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 75]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
43.  Forget MA, Turcotte S, Beauseigle D, Godin-Ethier J, Pelletier S, Martin J, Tanguay S, Lapointe R. The Wnt pathway regulator DKK1 is preferentially expressed in hormone-resistant breast tumours and in some common cancer types. Br J Cancer. 2007;96:646-653.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 124]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
44.  Qian J, Xie J, Hong S, Yang J, Zhang L, Han X, Wang M, Zhan F, Shaughnessy JD, Epstein J. Dickkopf-1 (DKK1) is a widely expressed and potent tumor-associated antigen in multiple myeloma. Blood. 2007;110:1587-1594.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 91]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
45.  Hall CL, Bafico A, Dai J, Aaronson SA, Keller ET. Prostate cancer cells promote osteoblastic bone metastases through Wnts. Cancer Res. 2005;65:7554-7560.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 220]  [Cited by in F6Publishing: 213]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
46.  Thudi NK, Martin CK, Nadella MV, Fernandez SA, Werbeck JL, Pinzone JJ, Rosol TJ. Zoledronic acid decreased osteolysis but not bone metastasis in a nude mouse model of canine prostate cancer with mixed bone lesions. Prostate. 2008;68:1116-1125.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 41]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
47.  Schwaninger R, Rentsch CA, Wetterwald A, van der Horst G, van Bezooijen RL, van der Pluijm G, Löwik CW, Ackermann K, Pyerin W, Hamdy FC. Lack of noggin expression by cancer cells is a determinant of the osteoblast response in bone metastases. Am J Pathol. 2007;170:160-175.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 75]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
48.  Voorzanger-Rousselot N, Goehrig D, Journe F, Doriath V, Body JJ, Clézardin P, Garnero P. Increased Dickkopf-1 expression in breast cancer bone metastases. Br J Cancer. 2007;97:964-970.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 145]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
49.  Yamabuki T, Takano A, Hayama S, Ishikawa N, Kato T, Miyamoto M, Ito T, Ito H, Miyagi Y, Nakayama H. Dikkopf-1 as a novel serologic and prognostic biomarker for lung and esophageal carcinomas. Cancer Res. 2007;67:2517-2525.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 159]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
50.  Uhlar CM, Whitehead AS. Serum amyloid A, the major vertebrate acute-phase reactant. Eur J Biochem. 1999;265:501-523.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 783]  [Cited by in F6Publishing: 767]  [Article Influence: 30.7]  [Reference Citation Analysis (0)]
51.  de Beer MC, de Beer FC, Gerardot CJ, Cecil DR, Webb NR, Goodson ML, Kindy MS. Structure of the mouse Saa4 gene and its linkage to the serum amyloid A gene family. Genomics. 1996;34:139-142.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 24]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
52.  Diamandis EP. Analysis of serum proteomic patterns for early cancer diagnosis: drawing attention to potential problems. J Natl Cancer Inst. 2004;96:353-356.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 335]  [Cited by in F6Publishing: 337]  [Article Influence: 16.9]  [Reference Citation Analysis (0)]
53.  Diamandis EP. Point: Proteomic patterns in biological fluids: do they represent the future of cancer diagnostics? Clin Chem. 2003;49:1272-1275.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 261]  [Cited by in F6Publishing: 231]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
54.  Akashi H, Hinoda Y, Itoh F, Adachi M, Endo T, Imai K. A novel gastric-cancer-associated mucin antigen defined by a monoclonal antibody A3D4. Int J Cancer. 1997;73:795-801.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
55.  Ren J, Chen Z, Juan SJ, Yong XY, Pan BR, Fan DM. Detection of circulating gastric carcinoma-associated antigen MG7-Ag in human sera using an established single determinant immuno-polymerase chain reaction technique. Cancer. 2000;88:280-285.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 3]  [Reference Citation Analysis (0)]
56.  Fan D, Zhang X, Chen X, Mou Z, Hu J, Zhou S, Ding J, Wu K. Bird’s-eye view on gastric cancer research of the past 25 years. J Gastroenterol Hepatol. 2005;20:360-365.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 40]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
57.  Pals G, Azuma T, Mohandas TK, Bell GI, Bacon J, Samloff IM, Walz DA, Barr PJ, Taggart RT. Human pepsinogen C (progastricsin) polymorphism: evidence for a single locus located at 6p21.1-pter. Genomics. 1989;4:137-148.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 27]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
58.  Mitri Z, Constantine T, O’Regan R. The HER2 Receptor in Breast Cancer: Pathophysiology, Clinical Use, and New Advances in Therapy. Chemother Res Pract. 2012;2012:743193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 138]  [Cited by in F6Publishing: 217]  [Article Influence: 18.1]  [Reference Citation Analysis (0)]
59.  Olayioye MA. Update on HER-2 as a target for cancer therapy: intracellular signaling pathways of ErbB2/HER-2 and family members. Breast Cancer Res. 2001;3:385-389.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 184]  [Cited by in F6Publishing: 197]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
60.  Burstein HJ. The distinctive nature of HER2-positive breast cancers. N Engl J Med. 2005;353:1652-1654.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 357]  [Cited by in F6Publishing: 355]  [Article Influence: 18.7]  [Reference Citation Analysis (0)]
61.  Tan M, Yu D. Molecular mechanisms of erbB2-mediated breast cancer chemoresistance. Adv Exp Med Biol. 2007;608:119-129.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 101]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
62.  Santin AD, Bellone S, Roman JJ, McKenney JK, Pecorelli S. Trastuzumab treatment in patients with advanced or recurrent endometrial carcinoma overexpressing HER2/neu. Int J Gynaecol Obstet. 2008;102:128-131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 115]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
63.  Kaufmann R, Müller P, Hildenbrand G, Hausmann M, Cremer C. Analysis of Her2/neu membrane protein clusters in different types of breast cancer cells using localization microscopy. J Microsc. 2011;242:46-54.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 71]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
64.  Shah DK, Menon KM, Cabrera LM, Vahratian A, Kavoussi SK, Lebovic DI. Thiazolidinediones decrease vascular endothelial growth factor (VEGF) production by human luteinized granulosa cells in vitro. Fertil Steril. 2010;93:2042-2047.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 16]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
65.  Nagy JA, Feng D, Vasile E, Wong WH, Shih SC, Dvorak AM, Dvorak HF. Permeability properties of tumor surrogate blood vessels induced by VEGF-A. Lab Invest. 2006;86:767-780.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 90]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
66.  Murat A, Migliavacca E, Hussain SF, Heimberger AB, Desbaillets I, Hamou MF, Rüegg C, Stupp R, Delorenzi M, Hegi ME. Modulation of angiogenic and inflammatory response in glioblastoma by hypoxia. PLoS One. 2009;4:e5947.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 84]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
67.  Pinto MP, Badtke MM, Dudevoir ML, Harrell JC, Jacobsen BM, Horwitz KB. Vascular endothelial growth factor secreted by activated stroma enhances angiogenesis and hormone-independent growth of estrogen receptor-positive breast cancer. Cancer Res. 2010;70:2655-2664.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 40]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
68.  Wang S, Li X, Parra M, Verdin E, Bassel-Duby R, Olson EN. Control of endothelial cell proliferation and migration by VEGF signaling to histone deacetylase 7. Proc Natl Acad Sci USA. 2008;105:7738-7743.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 171]  [Cited by in F6Publishing: 182]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
69.  Ding XW, Yang WB, Gao S, Wang W, Li Z, Hu WM, Li JJ, Luo HS. Prognostic significance of hERG1 expression in gastric cancer. Dig Dis Sci. 2010;55:1004-1010.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 28]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
70.  Chen X, Johns DC, Geiman DE, Marban E, Dang DT, Hamlin G, Sun R, Yang VW. Krüppel-like factor 4 (gut-enriched Krüppel-like factor) inhibits cell proliferation by blocking G1/S progression of the cell cycle. J Biol Chem. 2001;276:30423-30428.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 194]  [Cited by in F6Publishing: 200]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
71.  Chen C, Bhalala HV, Qiao H, Dong JT. A possible tumor suppressor role of the KLF5 transcription factor in human breast cancer. Oncogene. 2002;21:6567-6572.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 119]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
72.  Chen C, Bhalala HV, Vessella RL, Dong JT. KLF5 is frequently deleted and down-regulated but rarely mutated in prostate cancer. Prostate. 2003;55:81-88.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 115]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
73.  Bateman NW, Tan D, Pestell RG, Black JD, Black AR. Intestinal tumor progression is associated with altered function of KLF5. J Biol Chem. 2004;279:12093-12101.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 107]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
74.  Chen C, Benjamin MS, Sun X, Otto KB, Guo P, Dong XY, Bao Y, Zhou Z, Cheng X, Simons JW. KLF5 promotes cell proliferation and tumorigenesis through gene regulation and the TSU-Pr1 human bladder cancer cell line. Int J Cancer. 2006;118:1346-1355.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 120]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
75.  Pavan Kumar P, Purbey PK, Sinha CK, Notani D, Limaye A, Jayani RS, Galande S. Phosphorylation of SATB1, a global gene regulator, acts as a molecular switch regulating its transcriptional activity in vivo. Mol Cell. 2006;22:231-243.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 127]  [Cited by in F6Publishing: 119]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
76.  Galande S, Purbey PK, Notani D, Kumar PP. The third dimension of gene regulation: organization of dynamic chromatin loopscape by SATB1. Curr Opin Genet Dev. 2007;17:408-414.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in F6Publishing: 114]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
77.  Yasui D, Miyano M, Cai S, Varga-Weisz P, Kohwi-Shigematsu T. SATB1 targets chromatin remodelling to regulate genes over long distances. Nature. 2002;419:641-645.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 387]  [Cited by in F6Publishing: 383]  [Article Influence: 17.4]  [Reference Citation Analysis (0)]
78.  Lu X, Cheng C, Zhu S, Yang Y, Zheng L, Wang G, Shu X, Wu K, Liu K, Tong Q. SATB1 is an independent prognostic marker for gastric cancer in a Chinese population. Oncol Rep. 2010;24:981-987.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
79.  Cotterman R, Jin VX, Krig SR, Lemen JM, Wey A, Farnham PJ, Knoepfler PS. N-Myc regulates a widespread euchromatic program in the human genome partially independent of its role as a classical transcription factor. Cancer Res. 2008;68:9654-9662.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 98]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
80.  Ross JS, Ali SM, Wang K, Palmer G, Yelensky R, Lipson D, Miller VA, Zajchowski D, Shawver LK, Stephens PJ. Comprehensive genomic profiling of epithelial ovarian cancer by next generation sequencing-based diagnostic assay reveals new routes to targeted therapies. Gynecol Oncol. 2013;130:554-559.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 61]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
81.  Opavský R, Pastoreková S, Zelník V, Gibadulinová A, Stanbridge EJ, Závada J, Kettmann R, Pastorek J. Human MN/CA9 gene, a novel member of the carbonic anhydrase family: structure and exon to protein domain relationships. Genomics. 1996;33:480-487.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 281]  [Cited by in F6Publishing: 272]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
82.  Nakagawa Y, Uemura H, Hirao Y, Yoshida K, Saga S, Yoshikawa K. Radiation hybrid mapping of the human MN/CA9 locus to chromosome band 9p12-p13. Genomics. 1998;53:118-119.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
83.  Kirkpatrick JP, Rabbani ZN, Bentley RC, Hardee ME, Karol S, Meyer J, Oosterwijk E, Havrilesky L, Secord AA, Vujaskovic Z. Elevated CAIX Expression is Associated with an Increased Risk of Distant Failure in Early-Stage Cervical Cancer. Biomark Insights. 2008;3:45-55.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
84.  Dominguez-Sola D, Ying CY, Grandori C, Ruggiero L, Chen B, Li M, Galloway DA, Gu W, Gautier J, Dalla-Favera R. Non-transcriptional control of DNA replication by c-Myc. Nature. 2007;448:445-451.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 469]  [Cited by in F6Publishing: 486]  [Article Influence: 28.6]  [Reference Citation Analysis (0)]
85.  Nie Z, Hu G, Wei G, Cui K, Yamane A, Resch W, Wang R, Green DR, Tessarollo L, Casellas R. c-Myc is a universal amplifier of expressed genes in lymphocytes and embryonic stem cells. Cell. 2012;151:68-79.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 757]  [Cited by in F6Publishing: 789]  [Article Influence: 65.8]  [Reference Citation Analysis (0)]
86.  Peng L, Seto E. Deacetylation of nonhistone proteins by HDACs and the implications in cancer. Handb Exp Pharmacol. 2011;206:39-56.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 73]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
87.  Choi YW, Bae SM, Kim YW, Lee HN, Kim YW, Park TC, Ro DY, Shin JC, Shin SJ, Seo JS. Gene expression profiles in squamous cell cervical carcinoma using array-based comparative genomic hybridization analysis. Int J Gynecol Cancer. 2007;17:687-696.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 85]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
88.  Minamiya Y, Ono T, Saito H, Takahashi N, Ito M, Mitsui M, Motoyama S, Ogawa J. Expression of histone deacetylase 1 correlates with a poor prognosis in patients with adenocarcinoma of the lung. Lung Cancer. 2011;74:300-304.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 104]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
89.  Zhang Z, Yamashita H, Toyama T, Sugiura H, Ando Y, Mita K, Hamaguchi M, Hara Y, Kobayashi S, Iwase H. Quantitation of HDAC1 mRNA expression in invasive carcinoma of the breast*. Breast Cancer Res Treat. 2005;94:11-16.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 213]  [Cited by in F6Publishing: 227]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
90.  Adams H, Fritzsche FR, Dirnhofer S, Kristiansen G, Tzankov A. Class I histone deacetylases 1, 2 and 3 are highly expressed in classical Hodgkin’s lymphoma. Expert Opin Ther Targets. 2010;14:577-584.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 62]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
91.  Fritzsche FR, Weichert W, Röske A, Gekeler V, Beckers T, Stephan C, Jung K, Scholman K, Denkert C, Dietel M. Class I histone deacetylases 1, 2 and 3 are highly expressed in renal cell cancer. BMC Cancer. 2008;8:381.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 119]  [Cited by in F6Publishing: 134]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
92.  Weichert W, Röske A, Niesporek S, Noske A, Buckendahl AC, Dietel M, Gekeler V, Boehm M, Beckers T, Denkert C. Class I histone deacetylase expression has independent prognostic impact in human colorectal cancer: specific role of class I histone deacetylases in vitro and in vivo. Clin Cancer Res. 2008;14:1669-1677.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 270]  [Cited by in F6Publishing: 278]  [Article Influence: 17.4]  [Reference Citation Analysis (0)]
93.  Cao L, Hu X, Zhang Y, Sun XT. Omental milky spots in screening gastric cancer stem cells. Neoplasma. 2011;58:20-26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 14]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
94.  Behrns KE. Gastric remnant cancer: Is elimination of the bug the answer?: Comment on “Screening of Helicobacter pylori infection after gastrectomy for cancer or peptic ulcer”. Arch Surg. 2010;145:967.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
95.  Song Xy, Zeng L, Jin W, Thompson J, Mizel DE, Lei K, Billinghurst RC, Poole AR, Wahl SM. Secretory leukocyte protease inhibitor suppresses the inflammation and joint damage of bacterial cell wall-induced arthritis. J Exp Med. 1999;190:535-542.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 114]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
96.  Bouchard D, Morisset D, Bourbonnais Y, Tremblay GM. Proteins with whey-acidic-protein motifs and cancer. Lancet Oncol. 2006;7:167-174.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 133]  [Cited by in F6Publishing: 142]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
97.  Devoogdt N, Revets H, Ghassabeh GH, De Baetselier P. Secretory leukocyte protease inhibitor in cancer development. Ann N Y Acad Sci. 2004;1028:380-389.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 41]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
98.  Thornalley PJ. Advances in glyoxalase research. Glyoxalase expression in malignancy, anti-proliferative effects of methylglyoxal, glyoxalase I inhibitor diesters and S-D-lactoylglutathione, and methylglyoxal-modified protein binding and endocytosis by the advanced glycation endproduct receptor. Crit Rev Oncol Hematol. 1995;20:99-128.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 85]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
99.  Zhang D, Tai LK, Wong LL, Chiu LL, Sethi SK, Koay ES. Proteomic study reveals that proteins involved in metabolic and detoxification pathways are highly expressed in HER-2/neu-positive breast cancer. Mol Cell Proteomics. 2005;4:1686-1696.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 212]  [Cited by in F6Publishing: 228]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
100.  Antognelli C, Del Buono C, Ludovini V, Gori S, Talesa VN, Crinò L, Barberini F, Rulli A. CYP17, GSTP1, PON1 and GLO1 gene polymorphisms as risk factors for breast cancer: an Italian case-control study. BMC Cancer. 2009;9:115.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 62]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
101.  Romanuik TL, Ueda T, Le N, Haile S, Yong TM, Thomson T, Vessella RL, Sadar MD. Novel biomarkers for prostate cancer including noncoding transcripts. Am J Pathol. 2009;175:2264-2276.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 86]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
102.  Bair WB, Cabello CM, Uchida K, Bause AS, Wondrak GT. GLO1 overexpression in human malignant melanoma. Melanoma Res. 2010;20:85-96.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 71]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
103.  Thornalley PJ, Rabbani N. Glyoxalase in tumourigenesis and multidrug resistance. Semin Cell Dev Biol. 2011;22:318-325.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 125]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
104.  Scholzen T, Gerdes J. The Ki-67 protein: from the known and the unknown. J Cell Physiol. 2000;182:311-322.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 48]  [Reference Citation Analysis (0)]
105.  Blobe GC, Schiemann WP, Lodish HF. Role of transforming growth factor beta in human disease. N Engl J Med. 2000;342:1350-1358.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1834]  [Cited by in F6Publishing: 1831]  [Article Influence: 76.3]  [Reference Citation Analysis (0)]
106.  Reiss M. TGF-beta and cancer. Microbes Infect. 1999;1:1327-1347.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 106]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
107.  Schmidt A, Langbein L, Prätzel S, Rode M, Rackwitz HR, Franke WW. Plakophilin 3--a novel cell-type-specific desmosomal plaque protein. Differentiation. 1999;64:291-306.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 41]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
108.  Bonné S, Gilbert B, Hatzfeld M, Chen X, Green KJ, van Roy F. Defining desmosomal plakophilin-3 interactions. J Cell Biol. 2003;161:403-416.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 128]  [Cited by in F6Publishing: 127]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
109.  Papagerakis S, Shabana AH, Depondt J, Gehanno P, Forest N. Immunohistochemical localization of plakophilins (PKP1, PKP2, PKP3, and p0071) in primary oropharyngeal tumors: correlation with clinical parameters. Hum Pathol. 2003;34:565-572.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 55]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
110.  Satoh S, Daigo Y, Furukawa Y, Kato T, Miwa N, Nishiwaki T, Kawasoe T, Ishiguro H, Fujita M, Tokino T. AXIN1 mutations in hepatocellular carcinomas, and growth suppression in cancer cells by virus-mediated transfer of AXIN1. Nat Genet. 2000;24:245-250.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 703]  [Cited by in F6Publishing: 697]  [Article Influence: 29.0]  [Reference Citation Analysis (0)]
111.  Götte M, Kersting C, Radke I, Kiesel L, Wülfing P. An expression signature of syndecan-1 (CD138), E-cadherin and c-met is associated with factors of angiogenesis and lymphangiogenesis in ductal breast carcinoma in situ. Breast Cancer Res. 2007;9:R8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 87]  [Article Influence: 5.1]  [Reference Citation Analysis (1)]
112.  Zabrenetzky V, Harris CC, Steeg PS, Roberts DD. Expression of the extracellular matrix molecule thrombospondin inversely correlates with malignant progression in melanoma, lung and breast carcinoma cell lines. Int J Cancer. 1994;59:191-195.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 150]  [Cited by in F6Publishing: 168]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
113.  Zajchowski DA, Band V, Trask DK, Kling D, Connolly JL, Sager R. Suppression of tumor-forming ability and related traits in MCF-7 human breast cancer cells by fusion with immortal mammary epithelial cells. Proc Natl Acad Sci USA. 1990;87:2314-2318.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 56]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
114.  Volpert OV, Stellmach V, Bouck N. The modulation of thrombospondin and other naturally occurring inhibitors of angiogenesis during tumor progression. Breast Cancer Res Treat. 1995;36:119-126.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 79]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
115.  Sheibani N, Frazier WA. Thrombospondin 1 expression in transformed endothelial cells restores a normal phenotype and suppresses their tumorigenesis. Proc Natl Acad Sci USA. 1995;92:6788-6792.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in F6Publishing: 132]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
116.  Bornstein P, Sage EH. Matricellular proteins: extracellular modulators of cell function. Curr Opin Cell Biol. 2002;14:608-616.  [PubMed]  [DOI]  [Cited in This Article: ]
117.  Brekken RA, Sage EH. SPARC, a matricellular protein: at the crossroads of cell-matrix communication. Matrix Biol. 2001;19:816-827.  [PubMed]  [DOI]  [Cited in This Article: ]
118.  Briggs J, Chamboredon S, Castellazzi M, Kerry JA, Bos TJ. Transcriptional upregulation of SPARC, in response to c-Jun overexpression, contributes to increased motility and invasion of MCF7 breast cancer cells. Oncogene. 2002;21:7077-7091.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 86]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
119.  Ledda F, Bravo AI, Adris S, Bover L, Mordoh J, Podhajcer OL. The expression of the secreted protein acidic and rich in cysteine (SPARC) is associated with the neoplastic progression of human melanoma. J Invest Dermatol. 1997;108:210-214.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 127]  [Cited by in F6Publishing: 131]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
120.  Wang CS, Lin KH, Chen SL, Chan YF, Hsueh S. Overexpression of SPARC gene in human gastric carcinoma and its clinic-pathologic significance. Br J Cancer. 2004;91:1924-1930.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 96]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
121.  Otake Y, Soundararajan S, Sengupta TK, Kio EA, Smith JC, Pineda-Roman M, Stuart RK, Spicer EK, Fernandes DJ. Overexpression of nucleolin in chronic lymphocytic leukemia cells induces stabilization of bcl2 mRNA. Blood. 2007;109:3069-3075.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 178]  [Cited by in F6Publishing: 171]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
122.  Ye T, Chen Y, Fang J. DNA methylation biomarkers in serum for gastric cancer screening. Mini Rev Med Chem. 2010;10:1034-1038.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
123.  Correa P. Serum pepsinogens in gastric cancer screening. Dig Dis Sci. 2010;55:2123-2125.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 18]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
124.  Nakashima H, Nagahama R, Yamamoto T, Ohkura Y. Mass screening for gastric cancer: how to select patients for endoscopic examination. Gastric Cancer. 2010;13:78-83.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
125.  Suzuki H, Gotoda T, Sasako M, Saito D. Detection of early gastric cancer: misunderstanding the role of mass screening. Gastric Cancer. 2006;9:315-319.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 54]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
126.  Sako A, Kitayama J, Ishikawa M, Yamashita H, Nagawa H. Impact of immunohistochemically identified lymphatic invasion on nodal metastasis in early gastric cancer. Gastric Cancer. 2006;9:295-302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 36]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
127.  Oda I, Saito D, Tada M, Iishi H, Tanabe S, Oyama T, Doi T, Otani Y, Fujisaki J, Ajioka Y. A multicenter retrospective study of endoscopic resection for early gastric cancer. Gastric Cancer. 2006;9:262-270.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 291]  [Cited by in F6Publishing: 317]  [Article Influence: 18.6]  [Reference Citation Analysis (0)]
128.  Hasuo T, Semba S, Li D, Omori Y, Shirasaka D, Aoyama N, Yokozaki H. Assessment of microsatellite instability status for the prediction of metachronous recurrence after initial endoscopic submucosal dissection for early gastric cancer. Br J Cancer. 2007;96:89-94.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 20]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
129.  Tsai MM, Lin PY, Cheng WL, Tsai CY, Chi HC, Chen CY, Tseng YH, Cheng YF, Chen CD, Liang Y. Overexpression of ADP-ribosylation factor 1 in human gastric carcinoma and its clinicopathological significance. Cancer Sci. 2012;103:1136-1144.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 23]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]