Protein Signatures for Classification and Prognosis of Gastric Cancer
Daguang Wang,Fei Ye,Yabin Sun,Wei Li,Hongyi Liu,Jing Jiang,Yang Zhang,Chengkui Liu,Wei Tong,Ling Gao,Yunguang Sun,Weijia Zhang,Terry SeeToe,Peng Lee,Jian Suo,David Y. Zhang
DOI: https://doi.org/10.1016/j.ajpath.2011.06.010
2011-01-01
Abstract:Current methods have limited accuracy in predicting survival and stratifying patients with gastric cancer for appropriate treatment. We sought to identify protein signatures of gastric cancer for classification and prognostication. The Protein Pathway Array (initial study) and Western blot (confirmation) were used to assess the protein expression in a total of 199 fresh frozen gastric samples. There were 56 paired samples divided into a training set (n = 37) and a validation set (n = 19) for the identification of differentially expressed proteins between tumor and normal tissues. There were 56 tumor samples used to identify proteins correlating with tumor and nodal staging. All 93 tumor samples were used to identify candidate proteins for predicting survival. We confirmed the survival prediction of the candidate proteins by using an additional cohort of gastric cancer samples (n = 50). There were 22 proteins differentially expressed between normal and tumor tissues. Nine proteins were selected to build the predictor to classify normal and tumor samples. Ten proteins were differentially expressed among different T stages and four of these were associated with invasive behavior. An additional four proteins were associated with lymph node metastasis. Two proteins were identified as independent risk factors for overall survival. This study indicated that some dysregulated signaling proteins could be selected as useful biomarkers for tumor classification and predicting outcome in gastric cancer patients. Current methods have limited accuracy in predicting survival and stratifying patients with gastric cancer for appropriate treatment. We sought to identify protein signatures of gastric cancer for classification and prognostication. The Protein Pathway Array (initial study) and Western blot (confirmation) were used to assess the protein expression in a total of 199 fresh frozen gastric samples. There were 56 paired samples divided into a training set (n = 37) and a validation set (n = 19) for the identification of differentially expressed proteins between tumor and normal tissues. There were 56 tumor samples used to identify proteins correlating with tumor and nodal staging. All 93 tumor samples were used to identify candidate proteins for predicting survival. We confirmed the survival prediction of the candidate proteins by using an additional cohort of gastric cancer samples (n = 50). There were 22 proteins differentially expressed between normal and tumor tissues. Nine proteins were selected to build the predictor to classify normal and tumor samples. Ten proteins were differentially expressed among different T stages and four of these were associated with invasive behavior. An additional four proteins were associated with lymph node metastasis. Two proteins were identified as independent risk factors for overall survival. This study indicated that some dysregulated signaling proteins could be selected as useful biomarkers for tumor classification and predicting outcome in gastric cancer patients. Gastric cancer is the fourth most common malignancy and ranked as the second leading cause of cancer death worldwide.1Parkin D.M. Bray F. Ferlay J. Pisani P. Global cancer statistics, 2002.CA Cancer J Clin. 2005; 55: 74-108Crossref PubMed Scopus (17300) Google Scholar The geographic distribution of incidence and mortality of gastric cancer varies remarkably worldwide. Areas with high incidence include Japan, Korea, China, Eastern Europe, and parts of Latin America. The mortality of gastric cancer has declined in past decades, mainly due to early detection by gastric endoscopy.2Tsugane S. Sasazuki S. Diet and the risk of gastric cancer: review of epidemiological evidence.Gastric Cancer. 2007; 10: 75-83Crossref PubMed Scopus (347) Google Scholar However, unlike that of other common cancers, the prognosis for most gastric cancer is poor and has improved little for the past several decades. Despite recent advances in chemotherapy and surgical techniques, the overall 5-year survival rate is lower than 40%.3Jemal A. Siegel R. Ward E. Hao Y. Xu J. Murray T. Thun M.J. Cancer statistics, 2008.CA Cancer J Clin. 2008; 58: 71-96Crossref PubMed Scopus (10195) Google Scholar Perplexingly, the prognosis varies widely in patients with stage II or III disease for undetermined biological reasons. Currently, prognosis of gastric cancer is based on pathology (ie, histology type, invasion, and metastasis), radiological imaging (for staging), and other clinical factors (age and comorbidity), which all determine how patients should be managed (surgery and subsequent chemotherapy). However, these traditional clinicopathological factors have significant limitations. Therefore, a large effort has been made to search for molecular markers for diagnosis, classification, and prognosis of gastric cancer.4Mori M. Mimori K. Shiraishi T. Tanaka S. Ueo H. Sugimachi K. Akiyoshi T. p27 expression and gastric carcinoma.Nat Med. 1997; 3: 593Crossref PubMed Scopus (265) Google Scholar, 5Akama Y. Yasui W. Yokozaki H. Kuniyasu H. Kitahara K. Ishikawa T. Tahara E. Frequent amplification of the cyclin E gene in human gastric cancer.Jpn J Cancer Res. 1995; 86: 617-621Crossref PubMed Scopus (145) Google Scholar, 6Graziano F. Mandolesi A. Ruzzo A. Bearzi I. Testa E. Arduini F. Silva R. Muretto P. Mari D. Berardi R. Scartozzi M. Lai V. Cascinu S. Magnani M. Predictive and prognostic role of E-cadherin protein expression in patients with advanced gastric carcinomas treated with palliative chemotherapy.Tumour Biol. 2004; 25: 106-110Crossref PubMed Scopus (18) Google Scholar, 7Sanz-Ortega J. Steinburg S.M. Moro E. Saez M. Lopez J.A. Sierra E. Sanz-Esponera J. Merino M.J. Comparative study of tumor angiogenesis and immunohistochemistry for p53, c-erbB2, c-myc and EGFr as prognostic factors in gastric cancer.Histol Histopathol. 2000; 15: 455-462PubMed Google Scholar For example, cell cycle regulation factors (p27 and cyclin E),4Mori M. Mimori K. Shiraishi T. Tanaka S. Ueo H. Sugimachi K. Akiyoshi T. p27 expression and gastric carcinoma.Nat Med. 1997; 3: 593Crossref PubMed Scopus (265) Google Scholar, 5Akama Y. Yasui W. Yokozaki H. Kuniyasu H. Kitahara K. Ishikawa T. Tahara E. Frequent amplification of the cyclin E gene in human gastric cancer.Jpn J Cancer Res. 1995; 86: 617-621Crossref PubMed Scopus (145) Google Scholar cell adhesion molecules (E-cadherin),6Graziano F. Mandolesi A. Ruzzo A. Bearzi I. Testa E. Arduini F. Silva R. Muretto P. Mari D. Berardi R. Scartozzi M. Lai V. Cascinu S. Magnani M. Predictive and prognostic role of E-cadherin protein expression in patients with advanced gastric carcinomas treated with palliative chemotherapy.Tumour Biol. 2004; 25: 106-110Crossref PubMed Scopus (18) Google Scholar oncogenes (c-erbB2 and c-myc),7Sanz-Ortega J. Steinburg S.M. Moro E. Saez M. Lopez J.A. Sierra E. Sanz-Esponera J. Merino M.J. Comparative study of tumor angiogenesis and immunohistochemistry for p53, c-erbB2, c-myc and EGFr as prognostic factors in gastric cancer.Histol Histopathol. 2000; 15: 455-462PubMed Google Scholar and tumor suppressor genes (p53)7Sanz-Ortega J. Steinburg S.M. Moro E. Saez M. Lopez J.A. Sierra E. Sanz-Esponera J. Merino M.J. Comparative study of tumor angiogenesis and immunohistochemistry for p53, c-erbB2, c-myc and EGFr as prognostic factors in gastric cancer.Histol Histopathol. 2000; 15: 455-462PubMed Google Scholar have been reported to correlate with the prognosis of gastric cancer patients. Despite these reports, inconsistent results exist among the different studies, and the reported parameters provide limited information on the prognosis of individual patients because of the complex biology of the disease.8Zheng L. Wang L. Ajani J. Xie K. Molecular basis of gastric cancer development and progression.Gastric Cancer. 2004; 7: 61-77Crossref PubMed Scopus (163) Google Scholar In this study, we attempted to screen for proteins that can be used for diagnosis and prognosis of gastric cancer using the Protein Pathway Array method, a multiplex immunoblot-based assay combined with computational analysis.9Zhang D.Y. Ye F. Gao L. Liu X. Zhao X. Che Y. Wang H. Wang L. Wu J. Song D. Liu W. Xu H. Jiang B. Zhang W. Wang J. Lee P. Proteomics, pathway array and signaling network-based medicine in cancer.Cell Div. 2009; 4: 20-36Crossref PubMed Scopus (48) Google Scholar The Protein Pathway Array is a novel proteomic method that can characterize hundreds of proteins in clinical samples and identify alterations in protein expression or abundance with biomarker potential. We applied this unique approach to identify differentially expressed signal transduction proteins in gastric cancer tissue. Because the dysregulation of signal transduction proteins is responsible for cancer development, these proteins can be used as a signature for the diagnosis and prognosis of gastric cancer. Using this approach, we successfully identified a panel of nine proteins for distinguishing gastric cancer, four proteins associated with invasion, and two proteins for prognosis of survival. Fifty-six pairs of gastric cancer and adjacent nontumor mucosa (37 in the training set and 19 in the validation set), and an additional 87 cancer tissues (37 in the additional set and 50 in the second cohort) (Figure 1) were obtained after informed consent from patients who underwent D2 gastrectomy (ie, radical gastrectomy with level 2 extended lymphadenectomy) between February 2008 and June 2009 at The First Hospital of Jilin University, Jilin, China. This study was reviewed and approved by The First Hospital of Jilin University's Institution Ethical Review Boards. The representative tumors and adjacent normal tissues of these patients were dissected and frozen within 30 minutes of removal in a liquid nitrogen tank after immediate pathological examination. Tumor samples of 3 × 3 × 5 mm3 were taken from areas without gross necrosis. Adjacent nontumor mucosa samples of 3 × 3 × 5 mm3 were taken from the same patient at 3 cm away from the tumor margin. The tumor samples did not contain normal mucosal tissue, except for occasional entrapped gastric glands. The mucosa samples contained mucosa and a part of adherent submucosa; neither tumor nor dysplasia was included.9Zhang D.Y. Ye F. Gao L. Liu X. Zhao X. Che Y. Wang H. Wang L. Wu J. Song D. Liu W. Xu H. Jiang B. Zhang W. Wang J. Lee P. Proteomics, pathway array and signaling network-based medicine in cancer.Cell Div. 2009; 4: 20-36Crossref PubMed Scopus (48) Google Scholar The clinicopathological data of the patients are summarized in Table 1. A total of 143 patients (137 advanced and 6 early gastric cancers) were included (93 initial samples and 50 second cohort samples). One hundred and twenty patients had regional lymph node metastasis and one patient had distant metastasis (liver) at the surgery. The TNM stage of the tumor was done according to the American Joint Committee on Cancer.10Mullaney P.J. Wadley M.S. Hyde C. Wyatt J. Lawrence G. Hallissey M.T. Fielding J.W. Appraisal of compliance with the UICC/AJCC staging system in the staging of gastric cancer Union Internacional Contra la Cancrum/American Joint Committee on Cancer.Br J Surg. 2002; 89: 1405-1408Crossref PubMed Scopus (56) Google ScholarTable 1Patient Demographics and Gastric Cancer CharacteristicsClinicopathological characteristicsPatient number (%)First cohortSecond cohort(n = 93)(n = 50)Age ≤60 years38 (41)17 (34) >60 years55 (59)33 (66)Sex Male73 (78)24 (48) Female20 (22)26 (52)Family history Yes12 (13)13 (26) No81 (87)37 (74)Histology Histological grade Moderately differentiated adenocarcinoma30 (32)15 (30) Poorly differentiated adenocarcinoma63 (68)35 (70)Vascular invasion Yes60 (65)31 (62) No33 (35)19 (38)AJCC TNM stage⁎According to the American Joint Committee on Cancer (AJCC).10 I15 (16)0 (0) II16 (17)10 (20) III39 (42)39 (78) IV23 (25)1 (2)Primary tumor T16 (6)0 (0) T219 (20)0 (0) T364 (69)50 (100) T44 (4)0 (0)Node status N023 (24)0 (0) N126 (28)5 (10) N222 (24)35 (70) N322 (24)10 (20)Metastasis M092 (99)50 (100) M1†Metastasis to liver.1 (1)0 (0) According to the American Joint Committee on Cancer (AJCC).10Mullaney P.J. Wadley M.S. Hyde C. Wyatt J. Lawrence G. Hallissey M.T. Fielding J.W. Appraisal of compliance with the UICC/AJCC staging system in the staging of gastric cancer Union Internacional Contra la Cancrum/American Joint Committee on Cancer.Br J Surg. 2002; 89: 1405-1408Crossref PubMed Scopus (56) Google Scholar† Metastasis to liver. Open table in a new tab The proteins from 199 samples (56 paired samples and 87 unpaired tumors) were extracted, with 149 of them being used to assess the level of protein expression and phosphorylation using the Protein Pathway Array, and 50 of them being used to detect the expression levels of two candidate proteins using Western blot (Figure 1). Fifty six paired tumors and adjacent normal tissues were used to select the protein panel to distinguish between normal and tumor tissues of gastric cancer. These 56 pairs of samples were divided into a training set (n = 37) and a validation set (n = 19). A total of 56 tumor samples (including 19 tumor samples from the validation set and additional 37 new tumor samples) were used to identify the protein panel to distinguish different TNM stages. All 93 tumor samples were used to assess the candidate proteins for predicting survival. An additional cohort of gastric cancer samples (n = 50) were used to confirm the ability of candidate proteins to predict survival. Total proteins were extracted from the 149 fresh frozen gastric samples using 1× sample lysis buffer (Cell Signaling Technology, Danvers, MA) containing 20 mmol/L Tris-HCL (pH 7.5), 150 mmol/L NaCL, 1 mmol/L Na2EDTA, 1 mmol/L EGTA, 1% Triton, 2.5 mmol/L sodium pyrophosphate, 1 mmol/L β-glycerophosphate, 1 mmol/L Na3VO4, and 1 μg/mL leupeptin in the presence of 1× proteinase inhibitor cocktail (Roche Applied Science, Indianapolis, IN) and 1× phosphatase inhibitor cocktail (Roche Applied Science). The lysate was sonicated 3 times for 15 seconds each, and then centrifuged at 14,000 rpm for 30 minutes at 4°C. The protein concentration was determined with the BCA Protein Assay kit (Pierce, Rockford, IL). Approximately 300 μg of protein lysate was loaded in one well across the entire width of 10% SDS polyacrylamide and separated by electrophoresis, as previously described.11Ye F. Che Y. McMillen E. Gorski J. Brodman D. Saw D. Jiang B. Zhang D.Y. The effect of Scutellaria Baicalensis on the signaling network in hepatocellular carcinoma cells.Nutr Cancer. 2009; 61: 530-537Crossref PubMed Scopus (57) Google Scholar After electrophoresis, the proteins were transferred electrophoretically to a nitrocellulose membrane (Bio-Rad, Hercules, CA), which was then blocked for 1 hour with blocking buffer including either 5% milk or 3% bovine serum albumin in 1× Tris-HCI, NaCl, and Tween 20 (TBST) containing 20 mmol/L Tris-HCl (pH 7.5), 100 mmol/L NaCl, and 0.1% Tween-20. Next, the membrane was clamped on a Western blotting manifold (Mini-PROTEAN II Multiscreen Apparatus, Bio-Rad, Hercules, CA) that isolates 20 channels across the membrane. The multiplex immunoblot was performed using a total of 142 protein-specific or phosphorylation site-specific antibodies (Table 2). Four sets of antibodies (a total of 36 to 38 protein-specific or phosphorylation site-specific antibodies per set) were individually used for each membrane, and all of the antibodies (from various companies) were validated independently before inclusion in the Protein Pathway Array. For the first set of 36 primary antibodies, a mixture of two antibodies in the blocking buffer were added to each channel and then incubated at 4°C overnight. The membrane was then washed with 1× Tris-buffered saline and 1× TBST, and was further incubated with secondary anti-rabbit or anti-mouse antibody conjugated with horseradish peroxidase (Bio-Rad) for 1 hour at room temperature. The membrane was developed with chemiluminescence substrate (Immun-Star HRP Peroxide Buffer/Immun-Star HRP Luminol Enhancer, Bio-Rad), and chemiluminescent signals were captured using the ChemiDoc XRS System (Bio-Rad). The same membrane was then stripped off using stripping buffer (Restore Western Blot Stripping Buffer, Thermo Scientific, Rockford, IL) and then used to detect a second set of 36 primary antibodies as previously described. The signal of each protein were determined by densitometric scanning (Quantity One software package, Bio-Rad).Table 2List of Antibodies Included in the Protein Pathway ArrayAntibodies specific for phosphorylation p-PKCα(Ser657), p-EGFR (Tyr1068), p-HER2/ERBB2 (Tyr1221/1222), p-PDK1 (Ser241), p-PKCα/βII (Thr638/641), p-p53 (Ser392), p-Akt (Ser473), p-PTEN (Ser380), p-Rb (Ser780), p-survivin (Thr 34), p-beta-catenin (Ser33/37/Thr41), p-STAT5 (Tyr694), p-STAT3 (Ser727), p-ERK (Thr202/Tyr204), p-p70 S6 kinase (Thr389), p-VEGFR-2 (Tyr951), p-FGFR (Tyr653/654), p-EIF4B (Ser422), p-HGFR/C-Met (Y1234/Y1235), p-Smad (Ser463/465), p-ERK5 (Thr218/Tyr220), p-p90RSK (Ser380), p-CREB (Ser133), p-mTOR (Ser2448), p-PKCδ(Thr505), p-CDC2 (Tyr15), p-c-Jun (Ser73), p-SAPK/JNK (Thr183/Tyr185), p-FLT3 (Tyr 591), p-p38 (Thr180/Tyr182), p-GSK-3α/β(Ser21/9), p-FAK (Tyr397), p-RB (Ser807/811), p-HGFR/C-Met (Y1003).Antibodies for signal transduction proteins CyclinB1, cyclinD1, CDK6, CDC25B, cyclinE, CDK2, p27, BRCA1, CDK4, neu, 14-3-3 beta, cPKCα, ERK, EGFR, WEE1, CDC25C, HSP90, CHK1, MDM2, CDC2 p34, E2F-1, PCNA, c-myc, Notch1, beta-catenin, Akt, Trap, XIAP, Bcl-2, ETS1, HIF-1α, HIF-2α, TTF-1, p53, Notch4, PTEN, SRC-1, p300, c-Kit, Bax, N-cadherin, Raf-1, CDC42, EIF4B, TNF-α, vimentin, OPN, survivin, E-cadherin, TGF-β, p16, p27, WT1, Mesothelin, Cleaved Caspase-3, COX2, ATF-1, CREB, p21, NF-κB52, NF-κB50, calretinin, H-Ras, Bcl-6, K-Ras, alpha-tubulin, NF-κB p65, Myf-6, p15, ATR, Fas, SUMO-1, MetRS, Ep-CAM, FOXM1, Era, SYK, STAT1, Eg5, HIF-3α, RAD52, ATM, ABCG2, Bad-7, KLF6, CaMKKa, Topo IIa, p38, IL-1β, TERT, Ub, PR, Rap1, HCAM, Lyn, twist, TAP, patched, Erb,VEGF, GLI-3, FGF-7, p63, SK3, rhoB, WNT-1, TDP1, SLUG.Underlines indicate detectable expression in either tumor or normal tissues.All phosphorylation state-specific antibodies were obtained from Cell Signaling Technology (Danvers, MA), except p-HGFR/C-Met (Y1234/Y1235) and p-HGFR/C-Met (Y1003), which were purchased from R&D Systems (Minneapolis, MN).All non-phospho-antibodies were obtained from Santa Cruz Biotechnology (Santa Cruz, CA), except the following antibodies: 1) ERK, Akt, beta-catenin, Notch4, CREB, Cleaved Caspase-3, EIF4B, NF-κB52, NF-κB50, and STAT1 were obtained from Cell Signaling Technology (Danvers, MA); 2) XIAP was obtained from BD Biosciences (San Jose, CA); 3) TGF-β was obtained from R&D Systems (Minneapolis, MN).TFG, transforming growth factor. Open table in a new tab Underlines indicate detectable expression in either tumor or normal tissues. All phosphorylation state-specific antibodies were obtained from Cell Signaling Technology (Danvers, MA), except p-HGFR/C-Met (Y1234/Y1235) and p-HGFR/C-Met (Y1003), which were purchased from R&D Systems (Minneapolis, MN). All non-phospho-antibodies were obtained from Santa Cruz Biotechnology (Santa Cruz, CA), except the following antibodies: 1) ERK, Akt, beta-catenin, Notch4, CREB, Cleaved Caspase-3, EIF4B, NF-κB52, NF-κB50, and STAT1 were obtained from Cell Signaling Technology (Danvers, MA); 2) XIAP was obtained from BD Biosciences (San Jose, CA); 3) TGF-β was obtained from R&D Systems (Minneapolis, MN). TFG, transforming growth factor. The background was locally subtracted from raw protein signal and the background subtracted intensity was normalized by the “global median subtraction” normalization method to reduce the variations arising from experimental results derived from different runs (such as transferring and blotting efficiency, total protein loading amount, and exposure density). In detail, for each protein, its intensity was divided by total intensities of all proteins from each sample, and then multiplied by average intensities of all proteins in all samples. The normalized data were transformed to log2 and were used in the subsequent statistical analysis. Total proteins were extracted from 50 fresh frozen gastric cancer samples, as previously described. There were 20 μg of proteins that were fractionated by electrophoresis through a 10% SDS-polyacrylamide gel electrophoresis, and then the proteins were transferred onto a nitrocellulose membrane. The membrane was incubated with the primary antibodies, including Akt (1:1000 dilution) and cyclin-dependent kinase 2 (1:1000 dilution) (both from Santa Cruz Biotechnology, Santa Cruz, CA) at 4°C overnight. The membrane was then incubated with a secondary anti-rabbit antibody conjugated with horseradish peroxidase (Amersham, Arlington Height, IL). The protein was detected using chemiluminescence method and chemiluminescent signals were captured using the ChemiDoc XRS System (Bio-Rad), as previously described. The same membrane was then blotted using a monoclonal anti-β-actin antibody (1:10,000 dilution; Sigma, St. Louis, MO). The signal of each protein was determined by densitometric scanning (Quantity One software package, Bio-Rad). Paired Student's t-test and Significant Analysis of Microarray (SAM) tool (http://www-stat.stanford.edu/∼tibs/SAM) were used to select the proteins differentially expressed between tumors and normal tissues. K-fold cross validation (K = 10) was used to select those proteins with a great discriminating power to distinguish tumors from normal tissues. K-fold cross validation and unsupervised hierarchical clustering analysis were performed using BRB Array Tools software v.3.3.0 (http://linus.nci.nih.gov/BRB-ArrayTools.html). SPSS v.17.0 software (SPSS Inc., Chicago, IL) was used for Cox proportional hazard regression analysis to correlate the Protein Pathway Array data with the clinical data (TNM and survival), as well as for Kaplan-Meier and log-rank analysis of overall survival. There were 22 (of 142) proteins found to be differentially expressed between tumors and normal tissues in the training set (37 paired samples) using paired t-test and SAM analysis (P < 0.05 or q < 5%) (see Supplemental Table S1 at http://ajp.amjpathol.org) (Figure 2). Among them, 9 proteins and phosphoproteins were up-regulated in tumors, including proliferating cell nuclear antigen (PCNA), Notch4, CDK4, CDK6, XIAP, p-protein kinase C (PKC)α/βII, Akt, β-catenin, and p-PKCα, and 13 proteins were down-regulated in tumors, including p-ERK, cyclin B1, cyclin E, p27, E-cadherin, Hypoxia-inducible factor (HIF)-3α, Cdc25B, NF-κB52, TDP1, SK3, NF-κB50, SRC-1, and cyclin D1. To identify a robust set of proteins for classification, we carried out supervised K-fold cross validation (K = 10) using two class prediction models, including a support vector machine (SVM) and 3-nearest neighbor (3NN). Nine proteins (PCNA, Notch 4, p-ERK, CDK6, X-linked inhibitor of apoptosis (XIAP), CDK2, Akt, β-catenin, and NF-κB52) with the value of P < 0.01 were selected to build the SVM predictor. Five proteins (PCNA, Notch 4, p-ERK, CDK6, and XIAP) were selected to build the 3NN predictor. Ninety seven percent of the samples in the training set were correctly classified by either SVM or 3NN modeling. Only two samples (1 pair) in the training set were misclassified with this model. To further confirm the ability of these nine proteins to classify gastric cancer, we tested these proteins using a separate validation set of specimens (19 pairs) by 3NN and SVM models as previously described. All samples in the validation set were correctly classified by 3NN modeling (100% sensitivity and specificity), but 1 pair of the samples were misclassified by SVM modeling (95% sensitivity and specificity). A two-way hierarchical clustering analysis was performed for both sets of samples and revealed distinct patterns for both training sets (Figure 3A) and validation set (Figure 3B), although several samples were misclassified. We also compared the protein expression pattern between two histology grades (moderately and poorly differentiated tumors), and no significant difference was found.Figure 3Hierarchical clustering analysis of differentially expressed proteins in paired tumor and normal samples. The expression profile of nine proteins between the paired tumor and normal samples in training set (n = 37) (A) and validation set (n = 19) (B). The color scale showed the level of expression. Red indicates overexpression and green indicates underexpression, black indicates no change, and gray no expression. The number in each column represents the sample number. Each row represents a protein.View Large Image Figure ViewerDownload Hi-res image Download (PPT) To identify molecular markers to predict gastric cancer behaviors (ie, invasion and lymph node metastasis), we applied the SAM tool to identify proteins differentially expressed among different tumor groups. Based on the pathology report, we classified the level of tumor invasion into four (T stage) groups: 1) T1 stage group (mucosa/submucosa), 2) T2 stage group (muscularis propria/subserosa), 3) T3 stage group (serosa without invasion of adjacent structures), and 4) T4 stage group (adjacent structures). For the ability of lymph node metastasis, we classified the tumors into N0 (no lymph node involvement), N1 (≤6 positive nodes), N2 (7 to 15 positive nodes), and N3 (≥16 positive nodes) (N stage). Among different levels of invasion, 10 differentially expressed proteins were identified by SAM analysis (P < 0.05). Five proteins (E-cadherin, NFkB50, HIF-3α, cyclin B1, and cyclin E) were differentially expressed between T1 and T2, and 10 proteins (E-cadherin, β-catenin, NF-κB50, HIF-3α, cyclin B1, cyclin E, XIAP, TDP1, SK3, and CDC25B) were differentially expressed between T1 and T3, and T1 and T4, as well as between T1 and T2, T1 and T3, and T1 and T4. No differentially expressed proteins were identified between T2 and T3, and T2, and T4. Among these, 10 proteins (E-cadherin, beta-catenin, NF-κB50, HIF-3α, cyclin B1, cyclin E, XIAP, TDP1, SK3, and CDC25B) were differentially expressed between T1 and combined T3 and T4 as well as between T1 and combined T2, T3, and T4. No differentially expressed proteins were identified between T2 and combined T3 and T4. Among these 10 proteins, 4 proteins (E-cadherin, CDC25B, HIF-3α, and cyclin B1) were selected as the best predictors by K-fold cross-validation (K=10) analysis (with p<0.05) to distinguish T1 (early cancer) and combined T2, T3, and T4 (advanced cancer). Two-way hierarchical clustering analysis by BRB Array Tools software using these four proteins separated 56 tumors into two main groups: 23 tumor samples into group A and the 33 rest of the samples into group B (Figure 4). It is worthy to note that all six T1 tumors and six T2 tumors (of 12) were classified into group A and five of six T1 tumors were clustered into one subgroup. Twenty-six T3 and T4 tumors (78.8%) were clustered into group B, but only 11 T3 and T4 tumors (47.8%) were clustered into group A (χ2 = 5.796; P = 0.016). The results suggest that group A tumors represent a biologically less invasive cancer. Of these four proteins, two (cyclin B1 and CDC25B) were up-regulated and two (HIF-3α and E-cadherin) were down-regulated in group A tumors, suggesting these proteins are associated with invasive behavior of the gastric cancer. Among different N stages, four differentially expressed proteins were identified by SAM analysis (P < 0.05), including PCNA, NF-κB50, Notch 4, and CDK6. PCNA was down-regulated in N1 tumors when compared with N0 tumors. NF-κB50 was down-regulated in N2 tumors when compared with N1 tumors. Notch4 and CDK6 were down-regulated in N3 tumors and NF-κB50 was up-regulated in N3 tumors when compared with N2 tumors. These data suggest that these four proteins may be associated with lymph node metastasis. In addition, two proteins (HIF-3α and p-PKC α/β II) were found to be associated with vascular invasion of gastric cancer. Of these, HIF-3α was up-regulated in the tumors with vascular invasion (P = 0.042), whereas p-PKC α/β II was down-regulated in the tumors with vascular invasion (P = 0.042). To identify proteins that may predict overall survival, a univariate Cox proportion hazard regression analysis was performed on the 22 differentially expressed proteins in gastric cancer in a cohort of 93 patients (Table 1 and Figure 1). Two proteins (CDK2 and Akt) were found to correlate with overall survival with hazard ratios of 1.293 [P = 0.036; 95% confidence interval (CI): 1.017 to 1.644] and 1.431 (P = 0.028; 95% CI: 1.039 to 1.971), respectively. To determine whether these proteins can be independent prognostic markers, a multivariate analysis was performed taking into consideration other clinical parameters, such as age, sex, family history, histology grade, vascular invasion, and TNM stage (Table 3). The data showed that CDK2 and Akt still stood as independent predictors with hazard ratios of 1.289 (P = 0.044, 95% CI: 1.011 to 1.644) and 1.572 (P = 0.011, 95% CI: 1.111 to 2.224), respectively. In addition, age at surgery (P = 0.008) and TNM staging (P = 0.011) were also independent predictors of survival (Table 3). Based on the hierarchical clustering analysis of CDK2 and Akt expression, the tumor samples were separated into either high or low expression groups. The group with high level expression of CDK2 or Akt associated with a poorer prognosis according to Kaplan-Meier and log-rank survival analysis (P = 0.01 and P = 0.03, respectively
What problem does this paper attempt to address?
-
Proteomic and Metabolic Signaling Pathways in Different Prognosis of Gastric Cancer.
Guodong Lian,Daguang Wang,Chang Shu,Chengguo Wei,Fei Ye,David Y. Zhang,Jian Suo
DOI: https://doi.org/10.1200/jco.2014.32.15_suppl.e15087
IF: 45.3
2014-01-01
Journal of Clinical Oncology
Abstract:e15087 Background: Gastric cancer (GC) remains the fourth most common cancer and the second most common cause of cancer-related death worldwide. To tailor treatment regimens individually on the basis of patients’ chemosensitivity is a more effective strategy. Methods: After assessing the chemosensitivity status through survival time after surgery and adjuvant chemotherapy, 140 stage III (AJC) GC patients were split into two groups: chemosensitive (67 cases) and chemoresistant (73 cases). The differentially expressed proteins between those two groups were assessed using Protein Pathway Array (PPA). Data were analyzed by Significance Analysis of Microarrays (SAM) and BRB Array Tools software v.3.3.0. Ingenuity Pathway Analysis (IPA) was explored to further analysis the relevant signaling pathways. Results: Among 292 protein-specific or phosphorylation site-specific proteins tested, 147 were detected in cancer tissues. SAM analysis showed that 23 of these proteins/phosphorproteins had significant difference (q<5%) between chemosensitive and chemoresistant groups. We further carried out supervised K-fold cross validation (K=10) analysis using support vector machine (SVM), and 18 proteins (PLk1, FKHR, HDAC1, calpain 2, WT1, cdk4, β3 Tubulin, HMG-1, NMT1, Bcl-xL, V-ATPase H, tsg 101, calpastatin, P-cadherin, ADH, p-JNK, DACH1 and E-cadherin) were selected to separate gastric cancer of chemosensitive group from chemoresistant group with 94% correct rate, 94.8% sensitivity and 93% specificity. Six proteins (ERCC1, FKHR, PLK1, DACH1, E-Cadherin and ADH) were identified as independent predictors of chemoresistance. Using these 6 proteins, a risk score model was established to determine the risk of chemoresistance for each individual gastric cancer patient. Conclusions: Our current study indicated that some proteins differentially expressed in gastric cancer can be selected as clinically useful biomarkers. The risk score model is useful for determining patients’ risk of chemoresistance after radical surgery.
-
Protein Expression Profiles and Clinicopathologic Characteristics Associate with Gastric Cancer Survival
Wei Li,Yan Chen,Xuan Sun,Jupeng Yang,David Y. Zhang,Daguang Wang,Jian Suo
DOI: https://doi.org/10.1186/s40659-019-0249-0
2019-01-01
Biological Research
Abstract:Background Prognosis remains one of most crucial determinants of gastric cancer (GC) treatment, but current methods do not predict prognosis accurately. Identification of additional biomarkers is urgently required to identify patients at risk of poor prognoses. Methods Tissue microarrays were used to measure expression of nine GC-associated proteins in GC tissue and normal gastric tissue samples. Hierarchical cluster analysis of microarray data and feature selection for factors associated with survival were performed. Based on these data, prognostic scoring models were established to predict clinical outcomes. Finally, ingenuity pathway analysis (IPA) was used to identify a biological GC network. Results Eight proteins were upregulated in GC tissues versus normal gastric tissues. Hierarchical cluster analysis and feature selection showed that overall survival was worse in cyclin dependent kinase (CDK)2, Akt1, X-linked inhibitor of apoptosis protein (XIAP), Notch4, and phosphorylated (p)-protein kinase C (PKC) α/β2 immunopositive patients than in patients that were immunonegative for these proteins. Risk score models based on these five proteins and clinicopathological characteristics were established to determine prognoses of GC patients. These proteins were found to be involved in cancer related-signaling pathways and upstream regulators were identified. Conclusion This study identified proteins that can be used as clinical biomarkers and established a risk score model based on these proteins and clinicopathological characteristics to assess GC prognosis.
-
Identification of Key Genes and Signaling Pathways Associated with the Progression of Gastric Cancer
Chaoran Yu,Jie Chen,Junjun Ma,Lu Zang,Feng Dong,Jing Sun,Minhua Zheng
DOI: https://doi.org/10.1007/s12253-019-00781-3
2019-12-17
Abstract:Genomic features have been gradually regarded as part of the fundamentals to the clinical diagnosis and treatment for gastric cancer. However, the molecular alterations taking place during the progression of gastric cancer remain unclear. Therefore, identification of potential key genes and pathways in the gastric cancer progression is crucial to clinical practices. The gene expression profile, GSE103236, was retrieved for the identification of the differentially expressed genes (DEGs), followed by gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichments, gene set enrichment analysis (GSEA) and the protein-protein interaction (PPI) networks. Multiple bioinformatics platforms were employed for expression and prognostic analysis. Fresh frozen gastric cancer tissues were used for external validation. A total of 161 DEGs were identified from GSE103236. The PPI network-derived hub genes included collagen type I alpha 1 chain (COL1A1), tissue inhibitor of the metalloproteinases (TIMP1), Secreted Phosphoprotein 1 (SPP1), somatostatin (SST), neuropeptide Y (NPY), biglycan (BGN), matrix metallopeptidase 3 (MMP3), apolipoprotein E (APOE), ATPase H+/K+ transporting alpha subunit (ATP4A), lysyl oxidase (LOX). SPP1 (log rank p = 0.0048, HR = 1.39 [1.1–1.75]) and MMP3 (log rank p < 0.0001, HR = 1.77 [1.44–2.19]) were significantly associated with poor overall survival. Stage-specifically, both COL1A1 and BGN were correlated with significant in stage III and IV gastric cancer cases. LOX showed significant correlation with prognosis in stage I and stage II gastric cancer cases. Furthermore, cg00583003 of SPP1 and cg16466334 of MMP3 exhibited highly methylation level and significant prognostic values (SPP1: HR = 1.625, p = 0.013; MMP3: HR = 0.647, p = 0.011). Hub genes signature displayed a favorable prognostic value (p value = 5.227e-05). APOE demonstrated the highest correlation with CD8+ T cells, neutrophils, and dendritic cells whereas BGN had the highest correlation with macrophages. This study systematically explored the key genes and pathways involved in PGC and AGC, providing insights into therapeutic individualized management.
oncology,pathology
-
Differential Expression of COL1A1, COL1A2, COL6A3, and SULF1 as Prognostic Biomarkers in Gastric Cancer
Yan Hu,Jingjing Li,Haifeng Luo,Wenli Song,Jiyuan Yang
DOI: https://doi.org/10.2147/IJGM.S321265
IF: 2.145
2021-09-17
International Journal of General Medicine
Abstract:Yan Hu, Jingjing Li, Haifeng Luo, Wenli Song, Jiyuan Yang Department of Oncology, The First People's Hospital of Jingzhou, Jingzhou, Hubei, 434000, People's Republic of China Correspondence: Yan Hu Department of Oncology, The First People's Hospital of Jingzhou, Jingzhou, Hubei, 434000, People's Republic of China Tel +86 716 8513623 Email Background: Gastric cancer (GC) is among the most prevalent cancers globally. As such, there is a need to explore the mechanism underlying its pathogenesis and identify potential biomarkers for its prognosis. Methods: ONCOMINE was used to screen differentially expressed genes between GC and normal gastric mucosa. GEPIA was used to analyze the expression and correlation of candidate genes in tumor node metastasis (TNM) stage. STRING was used to construct protein interaction network. Kaplan–Meier plotter was used to analyze survival. TIMER was used to evaluate the association between candidate genes and immune cell infiltration. Results: From the ONCOMINE database, we found COL1A1, COL1A2, COL6A3, and SULF1 genes were significantly upregulated in stomach adenocarcinomas. There was a considerable correlation between the expression of COL1A1 (p = 0.029), COL1A2 (p = 0.004), COL6A3 (p = 0.002), SULF1 (p = 0.001), and the TNM stage. COL1A1 was positively correlated with ERBB2 (R = − 0.037, p = 0.46), while the other three genes were negatively correlated with ERBB2 (p > 0.05). The Kaplan–Meier plotter showed that low transcriptional levels of COL1A1 (p = 0.0020), COL1A2 (p = 0.0015), COL6A3 (p = 0.0015), and SULF1 (p = 0.0016) in gastric cancer patients were remarkably related to longer overall survival. In addition, there was a close relationship between chemokine expression and infiltration of the six immune cell types: B cells, macrophages, CD4+ T cells, CD8+ T cells, dendritic cells, and neutrophils, implying that the genes acted as indicators of both prognosis and immune status. Conclusion: Our findings implicate COL1A1, COL1A2, COL6A3, and SULF1 as candidate biomarkers for the prognosis of gastric cancer. Keywords: gastric cancer, biomarker, prognosis Gastric cancer (GC) is among the most prevalent cancers worldwide. The highest gastric cancer incidence rates occur in East Asia, South and Central America, and Eastern Europe. 1 Rates of GC are exceptionally high in Japan and Korea, where it is the most common cancer in men, and in China, where it is the most common cause of cancer-associated death. 2 Globally, there were about 1.03 million cases of GC, which led to the deaths of over 780,000 people in 2018, indicating that the disease is the fifth most prevalent and the third most common cause of cancer-associated deaths worldwide. 3,4 According to their anatomic location and histologic type, over 95% of GC are categorized as adenocarcinomas. 5 The main reason for the short overall survival (OS) and poor prognosis of gastric cancer patients is that the patients can not be diagnosed early. Advanced gastric cancer patients still account for a large proportion. Nowadays, various progressive treatment methods can not bring good curative effect for patients with advanced gastric cancer. Early diagnosis is still the primary and extremely important means to reduce the mortality of gastric cancer patients. So far, the diagnosis of gastric cancer in the world mainly depends on the pathological biopsy of gastroscope, but it is very difficult to carry out large-scale pathological biopsy. In addition, patients with gastric cancer have no specific clinical symptoms, especially early gastric cancer, which is easy to miss diagnosis. 6 The lack of sensitive and specific predictive factors for gastric cancer diagnosis is the fundamental reason why gastric cancer patients can not be diagnosed early. Therefore, it is very important to find high specificity and sensitivity biomarkers for early diagnosis and prognosis of gastric cancer. 7 Bioinformatics analysis, which can screen out the key genes related to diseases, has been widely used in clinical disease research. 8,9 How to further analyze the existing data resources with bioinformatics has become a research hotspot in cancer and bioinformatics. Herein, we employed many open databases to analyze the expression of COL1A1, COL1A2, COL6A3, and SULF1 in gastric cancer and assess their potential application as prognostic markers and therapeutic targets. Our results provide theoretical basis for further study of the molecular mechanism of gastric cancer and its diagnosis and prognosis. We obtained the microarrays data from the ONCOMINE database ( www.oncomine.org ) and identified five datasets involving COL1A1, COL1A2, COL6A3, and SULF1 after using the following filters: -Abstract Truncated-
medicine, general & internal
-
Overexpression of PhosphorylatedMammalianTarget of Rapamycin Predicts Lymph NodeMetastasis and Prognosis of Chinese Patients with Gastric Cancer
Guanzhen Yu,Jie-jun Wang,Ying Chen,Xi Wang,Jun Pan,Gang Li,ZhiliangJia,Qiang Li,James C. Yao,K. Xie
2009-01-01
Abstract:Purpose:We determined the expression of mammalian target of rapamycin (mTOR) and its activated form, p-mTOR, in Chinese patients with gastric cancer and its clinical effects and underlying mechanisms. Experimental Design:Tissue microarray blocks containing gastric cancer tissue and matched noncancer gastric tissue specimens obtained from1,072 patientswere constructed. Expressionof total mTOR and p-mTOR in these specimens was analyzed using immunohistochemical studies and confirmed byWestern blotting. Results: The overall rates of total mTOR and p-mTOR overexpression were 50.8% (545 of 1,072) and 46.5% (499 of 1,072), respectively. The p-mTOR overexpression was significantly correlated with total mTOR overexpression. Overexpression of total mTOR protein was significantly correlated with tumor differentiation, T1/T2 tumors, and stage I/II/III disease, whereas p-mTOR overexpression was significantly correlated with lymph node metastasis and all stage disease. The Cox proportional hazards model revealed that the overexpression of p-mTOR, but not total mTOR, was an independent prognostic factor for gastric cancer. The overexpression of p-mTOR also predicted the angiogenic phenotype of human gastric cancer and regulated angiogenesis of gastric cancer cells. Conclusions: Increased activation of mTOR is frequent in human gastric cancer and overexpression of p-mTOR is an independent prognostic factor, suggesting that mTOR pathway could be a potential target for therapy of this malignancy. Gastric cancer is the second leading cause of cancer-related deaths worldwide. More than one-third of all gastric cancer cases occur in China (1, 2). Researchers have described many biological markers associated with gastric cancer progression and outcome and observed differences in survival and clinicopathologic prognostic factors in patients with this disease in different countries and racial and ethnic groups within given populations and areas (3–8). Molecules involved in several signaling pathways were potential prognostic and therapeutic markers (9, 10). Determining the expression profiles of key molecules in the survival pathways in gastric cancer progression may aid in diagnosing and predicting disease progression. Because of their potential roles in gastric cancer progression, the phosphatidylinositol 3-kinase-dependent pathway and angiogenesis pathway have become the foci for development of novel anticancer drugs (9), including mammalian target of rapamycin (mTOR). The mTOR is a serine/threonine downstream mediator in phosphatidylinositol 3-kinase signaling pathway. Physiologically, mTOR is a central controller of eukaryotic cell growth and proliferation and plays a critical role in regulating important cellular functions, including proliferation, growth, survival, mobility, and angiogenesis (10, 11). Authors have reported activation of the mTOR pathway and overexpression of mTOR in several types of tumors, including hepatocellular (12), renal cell (13), prostate (14), and breast (15) cancers. Moreover, patients with breast cancer who had mTOR overexpression had a risk of recurrence three times greater than that in patients without mTOR overexpression (15). Lang et al. (16) recently reported overexpression of p-mTOR in 60% of intestinal-type and 64% of diffuse-type human gastric cancer cases and that Imaging, Diagnosis, Prognosis Authors’Affiliations: Department of Medical Oncology, Changzheng Hospital; Department of Pathology, Changhai Hospital, Shanghai, People’s Republic of China; Department of Dermatology and Skin Science, Jack Bell Research Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, British Columbia, Canada; and Departments of Gastrointestinal Medical Oncology and Cancer Biology,The University of Texas M. D. Anderson Cancer Center, Houston,Texas Received 8/18/08; revised 10/20/08; accepted 11/3/08; published OnlineFirst 02/17/2009. Grant support: Ministry of Science and Technology of the People’s Republic of China, National KeyTechnology R&DProgram grant 2006BAI02A05 (J.Wang and G.Yu) and America Cancer Society research scholar grant CSM-106640 (K. Xie). The costs of publication of this article were defrayed in part by the payment of page charges.This article must therefore be hereby marked advertisement in accordance with18 U.S.C. Section1734 solely to indicate this fact. Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). Requests for reprints: Jiejun Wang, Department of Medical Oncology, Changzheng Hospital, Hetian Road 64, Shanghai 200070, People’s Republic of China; Shanghai. Phone: 86-21-66540109-7045; Fax: 86-21-56327477; E-mail: jiejunw@csco.org.cn; or Keping Xie, Department of Gastrointestinal Medical Oncology,The University of Texas M. D. Anderson Cancer Center, Unit 426, 1515 Holcombe Boulevard, Houston,TX 77030. Phone: 713-792-2828; Fax: 713-7451163; E-mail: kepxie@mail.mdanderson.org. F2009 American Association for Cancer Research. doi:10.1158/1078-0432.CCR-08-2138 www.aacrjournals.org Clin Cancer Res 2009;15(5) March1, 2009 1821 Research. on April 11, 2017. © 2009 American Association for Cancer clincancerres.aacrjournals.org Downloaded from inhibition of expression of mTOR significantly impaired gastric cancer cell migration and proliferation. However, the role of mTOR activation in gastric cancer carcinogenesis and progression is poorly understood. The clinical significance of the expression mTOR protein and particularly the activated form of p-mTOR in patients with gastric cancer also remains unclear. Angiogenesis is a complex process and a crucial step in tumor formation and progression. This process is regulated by a balance between proangiogenic and antiangiogenic molecules. Among numerous proangiogenic factor, vascular endothelial growth factor (VEGF) has been established as the major angiogenic factor (17). Frequently observed in gastric cancer (18–20), overexpression of VEGF closely correlates with lymph node metastasis (21), liver metastasis (22), age (23), and tumor size (24) and often indicates a poor prognosis (25, 26) and therapy resistance (27). Although VEGF is constitutively expressed in many tumor cells, its expression is subject to several regulatory mechanisms (17). The potential role of mTOR pathway in VEGF regulation is supported by several recent studies, showing that inhibition of mTOR associated with a decreased VEGF expression (28–30). Given the critical role of VEGF in cancer angiogenesis in general, those recent findings suggest a critical link between mTOR activation and VEGF-mediated angiogenesis in gastric cancer. The present study sought to provide both clinical and experimental evidence for the significance of mTOR activation in gastric cancer and the potential underlying mechanisms of its effect on gastric cancer pathogenesis, particularly angiogenesis. Materials andMethods Patient specimens and tissue microarray construction. A total of 1,072 patients with gastric adenocarcinoma who underwent curative surgery without prior treatments at Changhai Hospital from 2001 to 2005 were enrolled in this study. Patients with other gastric tumors, such as neuroendocrine tumors, lymphoma, and sarcoma, were excluded from this study. The patients’ medical records were reviewed to obtain data including age at diagnosis, sex, tumor location, tumor size (diameter), nerve invasion, and American Joint Committee on Cancer stage. The mean age of patients at tumor resection was 59 years; 757 (71%) were male and 315 (29%) were female. Clinical follow-up results were available only for 669 patients from the Shanghai area (mean follow-up duration, 37 months; range, 21-73 months). Twentyone paraffin-embedded tissue microarray blocks of gastric tumor, matched normal gastric, and lymph node metastasis tissue specimens obtained from those patients were created using a manual arrayer (Beecher Instruments). Each block had two 1.5 mm cores of primary tumor tissue and at least one 1.5 mm core of matched nonneoplastic mucosal tissue. For patients with lymph node and/or liver metastasis, one or two 1.5 mm cores of metastatic tissue were included. Of the primary gastric cancer specimens, 57.6% (618 of 1,072 cases) were intestinal-type and 42.4% (454 of 1,072 cases) were diffuse-type according to the Lauren classification. Other patient characteristics are listed in Table 1. All of the tissue specimens were obtained for the present study with patient informed consent, and the use of the human specimens was approved by the Changhai Hospital Institutional Review Board. Immunostaining and evaluation. Consecutive sections (4 Am) of paraffin-embedded tissue microarrays blocks were prepared and processed for immunohistochemical analysis as described previously (7). Total mTOR, p-mTOR, and VEGF protein expression in the sections was detected with appropriate antibodies against total mTOR (dilution, 1:50; clone Y391; Abcam), p-mTOR (Ser; dilution, 1:100; clone 49F9; CST), and VEGF (dilution, 1:200; clone SP28; Lab Vision). Total mTOR, p-mTOR, and VEGF protein expression in those 1,072 cases was evaluated by two individuals using an Olympus CX31 microscope (Olympus Optical). A semiquantitative scoring system was used as described previously (7). Specifically, a underexpression was defined as no staining or staining positivity in tumor tissue being less than matched normal tissue, a normal expression as staining positivity being similar to matched normal tissue, and an overexpression as staining positivity being higher than normal tissue. Staining was scored independently by two individuals who were blinded to each other’s findings. All conflicting calls on scoring were adjudicated by a third individual. Quantification of tumor microvessel density. For CD34 staining, formalin-fixed, paraffin-embedded tumor tissue sections were processed and stained with a monoclonal goat anti-CD34 (PECAM
-
Protein Predictive Signatures for Lymph Node Metastasis of Gastric Cancer
W. Li,F. Ye,D. Wang,X. Sun,G. Lian,J. Jiang,J. Suo,D. Zhang
DOI: https://doi.org/10.1002/ijc.27864
2012-01-01
International Journal of Cancer
Abstract:Lymph node status remains one of most crucial indicators of gastric cancer prognosis and treatment planning. Current imaging methods have limited accuracy in predicting lymph node metastasis. We sought to identify protein markers in primary gastric cancer and to define a risk model to predict lymph node metastasis. The Protein Pathway Array (PPA) (initial selection) and Western blot (confirmation) were used to assess the protein expression in a total of 190 freshly frozen gastric cancer samples. The protein expression levels were compared between samples with lymph node metastasis (n = 73) and those without lymph node metastasis (n = 57) using PPA. There were 27 proteins differentially expressed between lymph node positive samples and lymph node negative samples. Five proteins (Factor XIII B, TFIIH p89, ADAM8, COX-2 and CUL-1) were identified as independent predictors of lymph node metastasis. Together with vascular/lymphatic invasion status, a risk score model was established to determine the risk of lymph node metastasis for each individual gastric cancer patient. The ability of this model to predict lymph node metastasis was further confirmed in a second cohort of gastric cancer patients (33 with and 27 without lymph node metastasis) using Western blot. This study indicated that some proteins differentially expressed in gastric cancer can be selected as clinically useful biomarkers. The risk score model is useful for determining patients' risk of lymph node metastasis and prognosis.
-
Establishment of Golgi apparatus-related genes signature to predict the prognosis and immunotherapy response in gastric cancer patients
Rui Liu,Weiwei Chu,Xiaojin Liu,Jie Hong,Haiming Wang
DOI: https://doi.org/10.1097/md.0000000000037439
IF: 1.6
2024-03-17
Medicine
Abstract:Gastric cancer, a prevalent malignant tumor worldwide and a leading cause of cancer-related mortality, poses a significant public health burden. In 2020 alone, the global incidence of gastric cancer exceeded 1 million cases, with over 750,000 deaths reported. [ 1 ] Stomach adenocarcinoma (STAD) represents approximately 95% of all gastric cancer cases. Despite notable advancements in treatment modalities, the overall 5-year survival rate for patients at an advanced stage remains dishearteningly low, hovering around 20%. [ 2 ] Even with radical resection and perioperative chemotherapy for locally advanced patients, the 5-year overall survival rate remains below 40%. [ 3–5 ] Accurate prognostic assessment plays a pivotal role in tailoring individualized treatment strategies for patients with gastric cancer. The current Lauren/World Health Organization classification and tumor-node-metastasis staging system constitute essential tools for selecting appropriate therapeutic interventions. [ 6 ] Nevertheless, existing prognostic models predominantly rely on conventional clinical and pathological features while neglecting crucial aspects such as tumor molecular characteristics and individual genetic variations. Consequently, there is an urgent need to develop an accurate, reliable, and predictive prognostic risk model specific to gastric cancer to inform treatment decisions effectively. In recent years, significant progress has been made in unraveling the molecular mechanisms underlying gastric cancer due to rapid advancements in high-throughput technologies and large-scale genomic projects like The Cancer Genome Atlas (TCGA). These endeavors have provided profound insights into the intricate interplay between tumor genomic abnormalities, epigenetic alterations, and immune-related gene dysregulation that drive gastric cancer development and progression. [ 7 , 8 ] Leveraging these novel biomarkers presents unprecedented opportunities for constructing robust prognostic risk models specifically tailored for gastric cancer.
medicine, general & internal
-
Clinical Significance of Fibrinogen and Platelet to Pre-Albumin Ratio in Predicting the Prognosis of Advanced Gastric Cancer
Huakai Tian,Zitao Liu,Zuo Zhang,Lipeng Zhang,Zhen Zong,Jiang Liu,Houqun Ying,Hui Li
DOI: https://doi.org/10.2147/jir.s412033
IF: 4.5
2023-10-03
Journal of Inflammation Research
Abstract:Huakai Tian, 1, &ast Zitao Liu, 2, &ast Zuo Zhang, 3, &ast Lipeng Zhang, 2, &ast Zhen Zong, 2 Jiang Liu, 2 Houqun Ying, 4 Hui Li 5 1 Department of General Surgery, First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China; 2 Department of Gastrointestinal Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China; 3 Department of Obstetrics and Gynecology, Second Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China; 4 Department of Nuclear Medicine, Jiangxi Province Key Laboratory of Laboratory Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China; 5 Department of Rheumatology and Immunology, First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China &astThese authors contributed equally to this work Correspondence: Hui Li, Department of Rheumatology and Immunology, First Affiliated Hospital of Nanchang University, 17 Yongwai Main Street, Nanchang, 330006, People's Republic of China, Tel +86-15079108591, Email Houqun Ying, Department of Nuclear Medicine, Jiangxi province Key Laboratory of Laboratory Medicine, the Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, People's Republic of China, Tel/Fax +86 791-86300410, Email Objective: The aim of this study was to investigate the clinical significance of Fibrinogen and Platelet to Pre-albumin Ratio(FPAR) in predicting the prognosis of patients with advanced gastric cancer(AGC) and to construct a predictive model. Methods: We collected clinical data from 489 postoperative patients with AGC. FPAR was divided into high and low groups according to the receiver operating characteristic (ROC) curve. The value of FPAR in predicting the prognosis of progressive gastric cancer was analysed using univariate and multivariable Cox regression analysis and its relationship with clinicopathological features. Finally, the Overall Survival(OS) and recurrence-free survival(RFS) prediction models were constructed and validated using FPAR. Results: Univariate and multifactorial cox regression analysis showed that grade (P< 0.001), TNM-stage (P< 0.001), chemotherapy (P< 0.001), and FPAR (OR=3.054,95% CI:2.088– 4.467, P< 0.001) were independent risk factors for OS; grade (P=0.021), N-stage (P=0.024), TNM-stage (P=0.033), and FPAR (OR=2.215,95% CI:1.634– 3.003, P< 0.001) were independent risk factors for RFS. Subgroup analysis showed that the FPAR-low group had higher OS and RFS than the FPAR-high group, regardless of the patient's TNM stage (p< 0.05). However, OS was instead higher in the the stage III-FPAR-low group than in the the stage II-FPAR-high group (p< 0.05), while RFS was not significantly different. Predictive models incorporating FPAR had better predictive performance than those without FPAR, showing wide range of net benefit and AUC. After correction, the 2-year AUC, 3-year AUC and C-index of the OS model were 0.737, 0.756, and 0.746; the 2-year AUC, 3-year AUC, and C-index of the RFS model were 0.738, 0.758, and 0.711. Conclusion: FPAR levels were associated with prognosis in patients with AGC and could independently predict RFS and OS. Keywords: advanced gastric cancer, FPAR, overall survival, recurrence-free survival, prognosis Gastric cancer (GC), ranking morbidity fifth and mortality third, is one of the most common malignant tumors of the digestive system. 1,2 In recent years, although the incidence of GC has declined, the mortality is on the rise. As we all know, except for early GC, the prognosis of AGC is poor. Although radical surgical resection combined with adjuvant therapy, including targeted, immune and other comprehensive treatments are currently used, the 5-year survival rate of AGC is still less than 50%. 3 It has been reported that over 70% of tumour-related deaths in patients with gastric cancer are related to tumour recurrence and metastasis. 4 Therefore, accurate judgment of the recurrence and metastasis risk and prognosis of gastric cancer patients will help guide the diagnosis and treatment plan. It is well known that the development of gastric cancer is an extremely complex process, closely related to systemic inflammation, immunity, nutritional status and other factors. 5 It has been shown that inflammation plays an important role in the tumour microenvironment and is closely related to the occurrence, development, infiltration and metastasis of tumors, and variou -Abstract Truncated-
immunology
-
Identification of Potential Biomarkers Associated with Prognosis in Gastric Cancer via Bioinformatics Analysis
Dong Li,Yi Yin,Muqun He,Jianfeng Wang
DOI: https://doi.org/10.12659/MSM.929104
2021-02-14
Abstract:BACKGROUND Gastric cancer (GC) is one of the leading causes of cancer-related mortality worldwide. We aimed to identify differentially expressed genes (DEGs) and their potential mechanisms associated with the prognosis of GC patients. MATERIAL AND METHODS This study was based on gene profiling information for 37 paired samples of GC and adjacent normal tissues from the GSE118916, GSE79973, and GSE19826 datasets in the Gene Expression Omnibus database. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were used to investigate the biological role of the DEGs. The protein-protein interaction (PPI) network was constructed by Cytoscape, and the Kaplan-Meier plotter was used for prognostic analysis. RESULTS We identified 119 DEGs, including 21 upregulated and 98 downregulated genes, in GC. The 21 upregulated genes were mainly enriched in extracellular matrix-receptor interaction, focal adhesion, and transforming growth factor-ß signaling, while the 98 downregulated genes were significantly associated with gastric acid secretion, retinol metabolism, and metabolism of xenobiotics by cytochrome P450. Thirty hub DEGs were obtained for further analysis. Twenty-five of the 30 hub DEGs were significantly associated with the prognosis of GC, and 21 of the 25 hub DEGs showed consistent expression trends within the 3 profile datasets. KEGG reanalysis of these 21 hub DEGs showed that COL1A1, COL1A2, COL2A1, COL11A1, THBS2, and SPP1 were mainly enriched in the extracellular matrix-receptor interaction pathways. CONCLUSIONS We identified 6 genes that were significantly related to the prognosis of GC patients. These genes and pathways could serve as potential prognostic markers and be used to develop treatments for GC patients.
-
Discovery of Prognostic Signature Genes for Overall Survival Prediction in Gastric Cancer
Changyuan Meng,Shusen Xia,Yi He,Xiaolong Tang,Guangjun Zhang,Tong Zhou
DOI: https://doi.org/10.1155/2020/5479279
IF: 2.809
2020-01-01
Computational and Mathematical Methods in Medicine
Abstract:Background. Gastric cancer (GC) is one of the most common malignant tumors in the digestive system with high mortality globally. However, the biomarkers that accurately predict the prognosis are still lacking. Therefore, it is important to screen for novel prognostic markers and therapeutic targets. Methods. We conducted differential expression analysis and survival analysis to screen out the prognostic genes. A stepwise method was employed to select a subset of genes in the multivariable Cox model. Overrepresentation enrichment analysis (ORA) was used to search for the pathways associated with poor prognosis. Results. In this study, we designed a seven-gene-signature-based Cox model to stratify the GC samples into high-risk and low-risk groups. The survival analysis revealed that the high-risk and low-risk groups exhibited significantly different prognostic outcomes in both the training and validation datasets. Specifically, CGB5, IGFBP1, OLFML2B, RAI14, SERPINE1, IQSEC2, and MPND were selected by the multivariable Cox model. Functionally, PI3K-Akt signaling pathway and platelet-derived growth factor receptor (PDGFR) were found to be hyperactive in the high-risk group. The multivariable Cox regression analysis revealed that the risk stratification based on the seven-gene-signature-based Cox model was independent of other prognostic factors such as TNM stages, age, and gender. Conclusion. In conclusion, we aimed at developing a model to predict the prognosis of gastric cancer. The predictive model could not only effectively predict the risk of GC but also be beneficial to the development of therapeutic strategies.
-
A Tumor Progression Related 7-Gene Signature Indicates Prognosis and Tumor Immune Characteristics of Gastric Cancer
Fen Liu,Zongcheng Yang,Lixin Zheng,Wei Shao,Xiujie Cui,Yue Wang,Jihui Jia,Yue Fu
DOI: https://doi.org/10.3389/fonc.2021.690129
IF: 4.7
2021-06-14
Frontiers in Oncology
Abstract:Background Gastric cancer is a common gastrointestinal malignancy. Since it is often diagnosed in the advanced stage, its mortality rate is high. Traditional therapies (such as continuous chemotherapy) are not satisfactory for advanced gastric cancer, but immunotherapy has shown great therapeutic potential. Gastric cancer has high molecular and phenotypic heterogeneity. New strategies for accurate prognostic evaluation and patient selection for immunotherapy are urgently needed. Methods Weighted gene coexpression network analysis (WGCNA) was used to identify hub genes related to gastric cancer progression. Based on the hub genes, the samples were divided into two subtypes by consensus clustering analysis. After obtaining the differentially expressed genes between the subtypes, a gastric cancer risk model was constructed through univariate Cox regression, least absolute shrinkage and selection operator (LASSO) regression and multivariate Cox regression analysis. The differences in prognosis, clinical features, tumor microenvironment (TME) components and immune characteristics were compared between subtypes and risk groups, and the connectivity map (CMap) database was applied to identify potential treatments for high-risk patients. Results WGCNA and screening revealed nine hub genes closely related to gastric cancer progression. Unsupervised clustering according to hub gene expression grouped gastric cancer patients into two subtypes related to disease progression, and these patients showed significant differences in prognoses, TME immune and stromal scores, and suppressive immune checkpoint expression. Based on the different expression patterns between the subtypes, we constructed a gastric cancer risk model and divided patients into a high-risk group and a low-risk group based on the risk score. High-risk patients had a poorer prognosis, higher TME immune/stromal scores, higher inhibitory immune checkpoint expression, and more immune characteristics suitable for immunotherapy. Multivariate Cox regression analysis including the age, stage and risk score indicated that the risk score can be used as an independent prognostic factor for gastric cancer. On the basis of the risk score, we constructed a nomogram that relatively accurately predicts gastric cancer patient prognoses and screened potential drugs for high-risk patients. Conclusions Our results suggest that the 7-gene signature related to tumor progression could predict the clinical prognosis and tumor immune characteristics of gastric cancer.
oncology
-
Clinical and prognostic significance of HIF-1α, PTEN, CD44v6, and survivin for gastric cancer: a meta-analysis.
Jing Chen,Tao Li,Qilun Liu,Haiyan Jiao,Wenjun Yang,Xiaoxia Liu,Zhenghao Huo
DOI: https://doi.org/10.1371/journal.pone.0091842
IF: 3.7
2014-01-01
PLoS ONE
Abstract:PURPOSE:This study was to quantitatively summarize published data for evaluating the clinical and prognostic significance of four proteins involved in hypoxia-inducible factor-1 (HIF-1α) regulation of the metastasis cascade.
METHODS:Searches were performed using the MEDLINE, EMBASE, Cochrane Library, and Chinese Biomedicine databases without any language restrictions. Studies were pooled and either the summary risk ratio (RR) or odds ratio (OR) was calculated. Potential sources of heterogeneity were sought out via subgroup and sensitivity analyses, and publication bias was also performed.
RESULTS:Seventeen studies evaluated HIF-1α, 20 studies evaluated phosphatase and tensin homolog (PTEN), 20 studies evaluated Survivin, and 16 studies evaluated CD44v6. Our results showed that increased HIF-1α expression was linked to a poor 5-year overall survival (RR = 1.508; 95% confidence interval (CI) 1.318-1.725; P<0.001). Decreased survival was heavily influenced by advanced tumor invasion (OR = 3.050; 95% CI 2.067-4.501; P<0.001), lymph node metastasis (1415 patients; OR = 3.486, 95% CI 2.737-4.440; P<0.001), distant metastasis (OR = 6.635; 95% CI 1.855-23.738; P = 0.004), vascular invasion (OR = 2.368; 95% CI 1.725-3.252; P<0.001), dedifferentiation (OR = 2.112; 95% CI 1.410-3.163; P<0.001), tumor size (OR = 1.921; 95% CI 1.395-2.647; P<0.001), and a higher TNM stage (OR = 2.762; 95% CI 1.941-3.942; P<0.001). Similarly, aberrant expression of PTEN, CD44v6, and Survivin were also observed in tumors that correlated with poor OS. The higher ORs of death at 5 years were 1.637 (95% CI = 1.452-1.845; P<0.001), 1.901 (95% CI = 1.432-2.525; P<0.001), and 1.627 (95% CI = 1.384-1.913; P<0.001), respectively, with an OR>2 for the main stratified meta-analyses of clinical factors.
CONCLUSIONS:Our findings indicate that HIF-1α/PTEN/CD44v6/Survivin, as measured by immunohistochemistry, can be used to predict the prognosis and potential for invasion and metastasis in Asian patients with gastric cancer. The development of strategies against this subset of proteins could lead to new therapeutic approaches.
-
Identification of a Novel Protein-Based Prognostic Model in Gastric Cancers.
Zhijuan Xiong,Chutian Xing,Ping Zhang,Yunlian Diao,Chenxi Guang,Ying,Wei Zhang
DOI: https://doi.org/10.3390/biomedicines11030983
IF: 4.757
2023-01-01
Biomedicines
Abstract:Gastric cancer (GC) is the third leading cause of cancer-related deaths worldwide. However, there are still no reliable biomarkers for the prognosis of this disease. This study aims to construct a robust protein-based prognostic prediction model for GC patients. The protein expression data and clinical information of GC patients were downloaded from the TCPA and TCGA databases, and the expressions of 218 proteins in 352 GC patients were analyzed using bioinformatics methods. Additionally, Kaplan–Meier (KM) survival analysis and univariate and multivariate Cox regression analysis were applied to screen the prognosis-related proteins for establishing the prognostic prediction risk model. Finally, five proteins, including NDRG1_pT346, SYK, P90RSK, TIGAR, and XBP1, were related to the risk prognosis of gastric cancer and were selected for model construction. Furthermore, a significant trend toward worse survival was found in the high-risk group (p = 1.495 × 10−7). The time-dependent ROC analysis indicated that the model had better specificity and sensitivity compared to the clinical features at 1, 2, and 3 years (AUC = 0.685, 0.673, and 0.665, respectively). Notably, the independent prognostic analysis results revealed that the model was an independent prognostic factor for GC patients. In conclusion, the robust protein-based model based on five proteins was established, and its potential benefits in the prognostic prediction of GC patients were demonstrated.
-
Relationship Between Four Tumor-Associated Bio-Markers and Prognosis of Gastric Cancer
Xuyang Wen,Qianwen Li,Yunxiang Du,Yan Shi,Jing Yuan,Li Chen,Qiong Sun,Guanghai Dai
DOI: https://doi.org/10.4314/tjpr.v16i6.6
2017-01-01
Tropical Journal of Pharmaceutical Research
Abstract:Purpose: To investigate the relationship between prognosis of gastric cancer (GC) and the expression of P53, Epidermal growth factor receptor (EGFR), Human epidermal growth factor receptor-2 (HER-2), and Vascular endothelial growth factor (VEGF).Methods: One hundred and forty-seven patients admitted to People's Liberation Army General Hospital (Beijing, China) with diagnosis of locally advanced GC were enrolled in the study. Follow-up data were obtained by outpatient review or telephone follow-up. Expressions of P53, EGFR, HER-2 and VEGF were determined by immunohistochemical staining. The relationship between protein expression, clinico-pathological factors, disease-free survival time (DFS) and overall survival (OS) were analyzed.Results: The expressions of EGER, HER-2, P53 and VEGF in GC were 17.7, 17.0, 41.0 and 55.9%, respectively. The expressions of EGFR and P53 were positively correlated (r = 0.306, p < 0.05), while the expressions of VEGF and HER-2 were negatively correlated (r = -0.2, p < 0.05). The expressions of EGFR, HER-2 and VEGF were not related to the clinico-pathological factors (p > 0.05) while expression of P53 was related only to histological grade (p < 0.05). Univariate analysis showed that OS and DFS were longer (p < 0.05) when P53 was lowly expressed. Multiple-factor analysis revealed that histological grade, infiltration depth and P53 expression were independent factors that influenced DFS.Conclusion: These results indicate that the expression of P53, EGFR, HER2 and VEGF can be used to predict prognosis of GC and screening of patients benefits from adjuvant chemotherapy.
-
Prognostic Biomarker in Advanced Gastric Cancer
Lan Mi,Xin Ji,Jiafu Ji
DOI: https://doi.org/10.3978/j.issn.2224-4778.2016.01.02
2016-01-01
Abstract:Gastric cancer is considered one of the most lethal tumors. Even with the decline in its incidence, the mortality rate of this disease has remained high-gastric cancer ranks third in terms of cancer-related death worldwide. Patient survival is highly dependent on the tumor stage at the time of diagnosis. Yet, gastric cancer is often either asymptomatic or causing only nonspecific symptoms in its early stages. By the time the symptoms occur, the cancer has usually reached an advanced stage. The current management for advanced gastric cancer (AGC) is a multidisciplinary approach; nevertheless, the prognosis of AGC is poor. The primary tumor (T), regional nodes (N), and metastasis (M) (TNM) classification, which was validated as the best predictor of patient survival, has limited power to fully reflect the prognosis. In the last decade, several biomarkers are identified to ameliorate the accuracy of patient prognosis and subsequent treatment decision-making. Undoubtedly, the discovery of novel molecular biomarkers and their establishment in clinical practice make sense in the era of "personalized" oncologic practice. The purpose of this review is to discuss the prognostic significance of the currently well-known biomarkers as well as to highlight some of the new candidate prognostic molecular markers. These prognostic markers include conventional tissue-based genetic and epigenetic alterations, noncoding RNAs, and proteins. We will also discuss the non-invasive biomarker with the ability to monitor real-time tumor dynamics, such as circulating tumor cells (CTCs) and cell-free nucleic acids (cfNAs): DNA, microRNA and long ncRNAs (lncRNAs).
-
The Prognostic Significance of Apoptosis-Related Biological Markers in Chinese Gastric Cancer Patients.
X. Liu,Y. Xu,Z. Long,H. Zhu,Y. Wang
DOI: https://doi.org/10.1200/jco.2011.29.4_suppl.108
IF: 45.3
2011-01-01
Journal of Clinical Oncology
Abstract:108 Background: The prognosis varied among the patients with the same stage, therefore there was a need for new prognostic and predictive factors. The aim of this study was to evaluate the relationship of apoptosis-related biological markers such as P21, P27, P53, Bcl-2, Bax, and c-myc, and clinicopathological features and their prognostic value.METHODSFrom January 1996 to December 2007, 4,426 patients had undergone gastrectomy for gastric cancer at Fudan University Shanghai Cancer Center. Among 501 patients, the expression levels of P21, P27, P53, Bcl-2, Bax, and c-myc were examined by immunohistochemistry. The prognostic value of biological markers and the correlation between biological markers and other clinicopathological factors were investigated.RESULTSThere were 339 males and 162 females (2.09:1) with a mean age of 57. The percentages of positive expression of P21, P27, P53, Bcl-2, Bax, and c-myc were 73%, 25%, 65%, 22%, 43%, and 58%, respectively. There was a strong correlation between P21, P53, Bax, and c-myc expression (p = 0.00). There was significant association between P27, Bcl-2, and Bax expression (p < 0.05). The P21 expression correlated with male (p = 0.00), histological grade (p = 0.00), Borrmann type (p = 0.02), tumor location (p = 0.01); the P53 expression with histological grade (p = 0.01); Bcl-2 expression with pathological stage (p = 0.01); Bax expression with male (p = 0.02), histological grade (p = 0.01), Borrmann type (p = 0.01), tumor location (p = 0.00), lymph node metastasis (p = 0.03), pathological stage (p = 0.01); c-myc expression with Borrmann type (p = 0.00). Bcl-2 expression was related with good survival in univariate analysis (p = 0.01). Multivariate analysis showed that Bcl-2 expression and pathological stage were defined as independent prognostic factors for gastric cancers. There was significant differences of overall 5-year survival rates according to Bcl-2 expression or not in stage III (p = 0.00).CONCLUSIONSThe expressionof Bcl-2 was an independent prognostic factor for Chinese patients with gastric cancer; it might be a candidate for the gastric cancer staging system. No significant financial relationships to disclose.
-
P-065Prognostic value of carbohydrate tumor markers and inflammation-based markers in gastric cancer
w kong,qishan wang,j wei,yuejin yang,yan zhang,zengda zou,bo liu
DOI: https://doi.org/10.1093/annonc/mdv233.65
IF: 51.769
2015-01-01
Annals of Oncology
Abstract:Introduction: Gastric cancer is the fourth most common cancer worldwide. In China, most cases are diagnosed as metastatic or recurrent gastric cancer (MRGC) with the5-year survival rate being less than 5%. It is important to obtain simple, feasible and cost-less markers to evaluate the prognosis of the MRGC patients.Methods: We investigated the correlation of tumor markers such as CEA, CA125, CA19-9, CA24-2, CA72-4 and inflammation-based factors such as mGPS, NLR and PLR with prognosis in 439 MRGC patients.Results: CA125 was more frequently positive with peritoneal recurrence, and CEA was more frequently positive in patients with liver metastases. In the univariate analysis of survival, the following variables were associated with shorter overall survival (OS): male, previous pathology such as nerves invasion and vessel invasion, elevated CEA, CA72-4, CA125 andCA19-9, and inflammation-based variables such as Alb, CRP, mGPS, PLR, NLR, Hb, LDH, AchE and AKP. In the multivariate analysis, mGPS, CEA and CA125 were independent prognostic factors for OS. An exploration of the potential prognostic index model including the three independent factors was carried out; MSTs for the low-, moderate- and high-risk groups were 12, 10.5 and 5 months.Conclusion: Elevated serum CEA, CA125 and mGPS inpatients with MRGC are independent negative predictor of prognosis. Introduction: Gastric cancer is the fourth most common cancer worldwide. In China, most cases are diagnosed as metastatic or recurrent gastric cancer (MRGC) with the5-year survival rate being less than 5%. It is important to obtain simple, feasible and cost-less markers to evaluate the prognosis of the MRGC patients. Methods: We investigated the correlation of tumor markers such as CEA, CA125, CA19-9, CA24-2, CA72-4 and inflammation-based factors such as mGPS, NLR and PLR with prognosis in 439 MRGC patients. Results: CA125 was more frequently positive with peritoneal recurrence, and CEA was more frequently positive in patients with liver metastases. In the univariate analysis of survival, the following variables were associated with shorter overall survival (OS): male, previous pathology such as nerves invasion and vessel invasion, elevated CEA, CA72-4, CA125 andCA19-9, and inflammation-based variables such as Alb, CRP, mGPS, PLR, NLR, Hb, LDH, AchE and AKP. In the multivariate analysis, mGPS, CEA and CA125 were independent prognostic factors for OS. An exploration of the potential prognostic index model including the three independent factors was carried out; MSTs for the low-, moderate- and high-risk groups were 12, 10.5 and 5 months. Conclusion: Elevated serum CEA, CA125 and mGPS inpatients with MRGC are independent negative predictor of prognosis.
-
Serum tumor markers (carcinoembryonic antigen, carbohydrate antigen 19-9, carbohydrate antigen 72-4, carbohydrate antigen 24-2, ferritin) and gastric cancer prognosis correlation
Jie-Wen Zhu,Ling-Zhen Gong,Qian-Wen Wang
DOI: https://doi.org/10.4240/wjgs.v16.i9.2808
2024-09-27
World J Gastrointest Surg
Abstract:Background: Gastric cancer is a kind of malignant tumor which is prevalent all over the world. Although some progress has been made in the treatment of gastric cancer, its prognosis is still not optimistic, so it is of great significance to find reliable prognostic indicators to guide the treatment and management of patients with gastric cancer. Aim: To explore the relationship between serum levels of five biomarkers [carcinoembryonic antigen (CEA), carbohydrate antigen (CA) 19-9, CA72-4, CA24-2, and ferritin] and prognosis in patients with gastric cancer. Methods: This study included 200 patients with gastric adenocarcinoma, and conducted an in-depth analysis of their baseline characteristics, relationship between tumor markers and staging, and prognosis. The study found that CA19-9 has a significant correlation with tumor stage, the average levels of CA24-2, CEA, CA72-4 and ferritin were slightly increased disregarding the stage of tumor. Survival analysis showed that increases in CEA, CA19-9, CA24-2, and ferritin were all associated with shortened overall survival of patients. Further multivariate analysis revealed that elevated serum CA72-4 levels were an independent adverse prognostic factor. Results: This study reveals that there is a significant correlation between the expression levels of serum tumor markers CEA, CA19-9, CA72-4, CA24-2 and ferritin in patients with gastric cancer and prognosis, and can be used as important indicators for prognostic evaluation of gastric cancer. In particular, markers that appear abnormally elevated initially may help identify gastric cancer patients with poor prognosis. Conclusion: Serum CEA and CA19-9 play an important role in the prognosis assessment of gastric cancer, and are effective tools to guide clinical practice and optimize individualized treatment strategies for gastric cancer patients.
-
Identification of Potential Key Genes Associated With the Pathogenesis and Prognosis of Gastric Cancer Based on Integrated Bioinformatics Analysis.
Xinkui Liu,Jiarui Wu,Dan Zhang,Zhitong Bing,Jinhui Tian,Mengwei Ni,Xiaomeng Zhang,Ziqi Meng,Shuyu Liu
DOI: https://doi.org/10.3389/fgene.2018.00265
IF: 3.7
2018-01-01
Frontiers in Genetics
Abstract:Background and Objective: Despite striking advances in multimodality management, gastric cancer (GC) remains the third cause of cancer mortality globally and identifying novel diagnostic and prognostic biomarkers is urgently demanded. The study aimed to identify potential key genes associated with the pathogenesis and prognosis of GC. Methods: Differentially expressed genes between GC and normal gastric tissue samples were screened by an integrated analysis of multiple gene expression profile datasets. Key genes related to the pathogenesis and prognosis of GC were identified by employing protein-protein interaction network and Cox proportional hazards model analyses. Results: We identified nine hub genes (TOP2A, COL1A1, COL1A2, NDC80, COL3A1, CDKN3, CEP55, TPX2, and TIMP1) which might be tightly correlated with the pathogenesis of GC. A prognostic gene signature consisted of CST2, AADAC, SERPINE1, COL8A1, SMPD3, ASPN, ITGBL1, MAP7D2, and PLEKHS1 was constructed with a good performance in predicting overall survivals. Conclusion: The findings of this study would provide some directive significance for further investigating the diagnostic and prognostic biomarkers to facilitate the molecular targeting therapy of GC.
-
Integrated Proteome and Phosphoproteome Analysis of Gastric Adenocarcinoma Reveals Molecular Signatures Capable of Stratifying Patient Outcome.
Xue Lu,Yunyun Fu,Lei Gu,Yunpeng Zhang,Antony Y. Liao,Tingting Wang,Bin Jia,Donglei Zhou,Lujian Liao
DOI: https://doi.org/10.1002/1878-0261.13361
2022-01-01
Molecular Oncology
Abstract:Metastasis is one of the main causes of low survival rate of gastric cancer patients. Exploring key proteins in the progression of gastric adenocarcinoma (GAC) may provide new candidates for prognostic biomarker development and therapeutic intervention. We applied quantitative mass spectrometry to compare the proteome and phosphoproteome of primary tumor tissues between GAC patients with and without lymph node metastasis (LNM). We then performed an integrated analysis of the proteomic and transcriptomic data to reveal the molecular features. We quantified a total of 5536 proteins, and we found 218 upregulated and 49 downregulated proteins in tumor samples from patients with LNM compared to those without LNM. Clustering analysis identified a number of hub proteins that have been previously shown to play important roles in gastric cancer progression. We also found that two extracellular proteins, TNXB and SPON1, are overexpressed in patients with LNM, which correlates with poor survival of GAC patients. Overexpression of TNXB and SPON1 was validated by western blotting and immunohistochemistry. Furthermore, treating gastric cancer cells with anti‐TNXB antibody significantly reduced cell migration. Finally, quantitative phosphoproteomic analysis combined with activity‐based kinase capture revealed a number of activated kinases in primary tumor tissues from patients with LNM, among which GSK3 might be a new target that warrants further study. Our study provides a snapshot of the proteome and phosphoproteome of GAC tumor tissues that have metastatic potential, and identifies potential biomarkers for GAC progression.