Open Access

A novel risk score model for stomach adenocarcinoma based on the expression levels of 10 genes

  • Authors:
    • Encui Guan
    • Feng Tian
    • Zhaoxia Liu
  • View Affiliations

  • Published online on: December 9, 2019     https://doi.org/10.3892/ol.2019.11190
  • Pages: 1351-1367
  • Copyright: © Guan et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Stomach adenocarcinoma (STAD) accounts for 95% of cases of malignant gastric cancer, which is the third leading cause of cancer‑associated mortality worldwide. The pathogenesis and effective diagnosis of STAD have become popular topics for research in the previous decade. In the present study, high‑throughput RNA sequencing expression profiles and clinical data from patients with STAD were obtained from The Cancer Genome Atlas database and were used as a training dataset to screen differentially expressed genes (DEGs). Prognostic DEGs were identified using univariate Cox regression analysis and were further screened by the least absolute shrinkage and selection operator regularization regression algorithm. The resulting genes were used to construct a risk score model, the validation and effectiveness evaluation of which were performed on an independent dataset downloaded from the Gene Expression Omnibus database. Stratified and functional pathway (gene set enrichment) analyses were performed on groups with different estimated prognosis. A total of 92 genes significantly associated with STAD prognosis were obtained by univariate Cox regression analysis, and 10 prognosis‑associated DEGs; hemoglobin b, chromosome 4 open reading frame 48, Dickkopf WNT signaling pathway inhibitor 1, coagulation factor V, serpin family E member 1, transmembrane protein 200A, NADPH oxidase organizer 1, C‑X‑C motif chemokine ligand 3, mannosidase endo‑α‑like and tripartite motif‑containing 31; were selected for the development of the risk score model. The reliability of this prognostic method was verified using a validation set, and the results of multivariate Cox analysis indicated that the risk score may serve as an independent prognostic factor. In functional DEG analysis, eight Kyoto Encyclopedia of Genes and Genomes pathways were identified to be significantly associated with STAD risk factors. Thus, the 10‑gene risk score model established in the present study was regarded as credible. This risk assessment tool may help identify patients with a high risk of STAD, and the proposed prognostic mRNAs may be useful in elucidating STAD pathogenesis.

Introduction

Gastric cancer (GC) is currently the fifth most common type of cancer and the third leading cause of cancer-associated mortality worldwide among both sexes (1,2). Stomach adenocarcinoma (STAD) accounts for 95% of malignant GC cases (3). The incidence of GC increased annually in young Hispanic and US populations (20–49 years) between 2,000 and 2014 (4,5). Patients with advanced GC exhibit poor prognosis, which is frequently explained by a lack of early diagnostic biomarkers and effective treatment (6). As the prognosis of GC is associated with the stage of the disease at diagnosis, novel effective diagnostic tools for early stages of GC are urgently required (7).

The phenotypic alterations and the molecular mechanisms underlying GC have been increasingly elucidated, and the majority of researchers believe that GC is a multifactorial disease, the development of which involves various risk factors, such as Helicobacter pylori infection, smoking habits and dietary factors (810). With the advances in molecular biology and genetic detection techniques, the aberrant expression of certain genes, including miR-125b, −199a and −100 has been demonstrated to be significantly associated with the pathogenesis and prognosis of GC (11). However, the aberrant expression of a limited number of genes cannot accurately reflect the pathogenesis and prognosis of GC. Therefore, it may be clinically useful to develop statistical models for disease risk prediction and tools for subsequent risk assessment (12,13).

Risk assessment tools are considered to be able to help estimate the probability that a person with a specific set of risk factors will develop a disease of interest (13). These risk assessment tools can facilitate the identification of high-risk populations in relation to a disease and are useful in the subsequent clinical decision-making process for healthcare providers and patients (12). Risk assessment tools have been used to predict the outcome of a number of diseases, such as thromboembolism, Lynch syndrome and certain types of cancer, including GC (12,1420). This indicates the potential to establish a risk assessment tool using valuable prognostic factors with predictive capacity. Wang et al (20) developed a 53-gene signature for predicting prognosis of patients with GC. Although the prognostic scoring system has been demonstrated to successfully predict patient overall survival, the detection of expression of these 53 genes in one patient at a time is a complicated task in the clinical setting. Therefore, further efforts to establish a prognostic prediction model with fewer genes are still warranted.

The present study aimed to use large amounts of mRNA expression profiling data from STAD samples to screen significantly differentially expressed genes (DEGs) and establish a risk score (RS) model based on the screened genes. The RS model was simultaneously validated by means of an independent dataset from another database and via a correlation analysis between clinical characteristics and prognosis. This RS model might provide a new tool for predicting the prognosis of patients with STAD.

Materials and methods

Analysis workflow

The steps of the workflow were as follows: i) High-throughput RNA sequencing (RNA-seq) expression profiles and clinical data from patients with STAD were downloaded from The Cancer Gene Atlas (TCGA) database (https://portal.gdc.cancer.gov) and to be used as a training dataset; ii) the samples in the training set were subdivided into tumor and control samples according to the clinical data and were subjected to screening to identify DEGs; iii) prognostic DEGs were identified in the training set by univariate Cox regression analysis; iv) the prognostic DEGs selected in the previous step were screened using the least absolute shrinkage and selection operator (LASSO) regularization regression algorithm (21), and the resulting genes were used to develop the RS model. The model validation and effectiveness evaluation were performed on an independent dataset retrieved from the Gene Expression Omnibus (GEO) database (http://www.ncbi.nlm.nih.gov/geo); v) screening and stratified analysis of clinical factors were performed to identify independent prognostic risk factors; vi) screening of mRNA-seq data for DEGs and Gene Set Enrichment Analysis (GSEA) were performed in groups with different estimated prognosis. The overall analysis process is presented in Fig. 1.

Data

On April 8, 2018, the training dataset, comprising 348 samples with RNA-seq expression profile data and corresponding clinical information, was downloaded from TCGA. The 348 samples comprised 37 control samples and 311 STAD samples with survival times >6 months. The validation dataset GSE62254 (22), comprising 300 samples with RNA-seq expression profile data and the corresponding clinical information, was retrieved from the National Center for Biotechnology Information GEO database [platform GPL570 (HG-U133_Plus_2), Affymetrix Human Genome U133 Plus 2.0 Array]. The clinical information from the training and validation datasets is presented in Table I.

Table I.

Clinical information of samples in the training and validation datasets.

Table I.

Clinical information of samples in the training and validation datasets.

CharacteristicTCGA (n=311)GSE62254 (n=300)
Age, years, mean ± SD,64.91±10.2361.94±11.36
Sex, male/female203/108199/101
Subtype, MSI-H/MSI-L/MSS/unknown60/42/208/1Unknown
Reflux, yes/no/unknown36/163/112Unknown
H. pylori infection, yes/no/unknown20/141/150Unknown
Pathological T, T1/T2/T3/T4/unknown15/64/138/93/12/186/91/21
Pathological N, N0/N1/N2/N393/82/60/70/638/131/80/51
Pathological M, M0/M1/unknown285/15/11273/27/0
Pathological stage, I/II/III/IV/unknown43/96/145/26/130/96/95/77/2
Grade, 1/2/3/unknown7/109/186/9Unknown
Antireflux treatment, yes/no/unknown30/137/144Unknown
Radiation therapy, yes/no/unknown57/248/6Unknown
Recurrence, yes/no44/218125/157/18
Survival status, dead/alive/unknown122/189/0135/148/17
Progression-free survival, months, mean ± SD17.78±17.2833.72±29.82
Overall survival, months, mean ± SD18.45±17.1550.59±31.42

[i] TCGA, The Cancer Genome Atlas; SD, standard deviation; MSI-H, microsatellite instability-high; MSI-L, microsatellite instability-low; MSS, microsatellite stability. Tumors were staged according to the 7th edition TNM staging system (55).

Screening of RNA-seq data for mRNAs with significant differential expression between groups with different prognosis

The edgeR package, version 3.20.9, of the R 3.4.0 language (http://bioconductor.org/packages/release/bioc/html/edgeR.html) (23) was used with the training dataset to identify significant differences in mRNA expression between the 311 STAD samples and 37 control samples. There were two thresholds selected: False discovery rate <0.05 and |log2(fold-change)| >0.5. According to the significant differences in mRNA expression between the training set samples, two-way hierarchical clustering analysis of the mRNA expression values was performed using pheatmap version 1.0.8 in R 3.4.0 (https://cran.r-project.org/web/packages/pheatmap/index.html) (24) according to a centered Pearson correlation algorithm (25).

Identification of prognosis-associated mRNAs and clinical factors

Cox regression analysis was performed on the 311 STAD tumor samples in the training dataset using survival package version 2.41.3 in R 3.4.0 (https://cran.r-project.org/web/packages/survival/index.html) (26,27) to screen the mRNAs and clinical factors for those significantly associated with overall survival time. The screening threshold was log-rank test P<0.05.

Establishment and evaluation of the risk assessment tool (RS model)
Selection of an optimal mRNA combination

Based on the identified prognosis-associated mRNAs, the optimal mRNA combination was identified by the LASSO regularization regression algorithm (21) in the penalized package, version 0.9.50 in R 3.4.0 (https://cran.r-project.org/web/packages/penalized/index.html) (28). Optimized parameter l in the screening model was determined through cyclic execution of 1,000 repetitions of cross-validation likelihood (CVL) algorithm (29).

Determination of the mRNA expression level cutoff

For each mRNA included in the optimal mRNA combination, the expression level cutoff value was determined using the X-Tile Software (https://medicine.yale.edu/lab/rimm/research/software.aspx) (30). Monte-Carlo P<0.05 was used as the threshold to determine the cut-off value. The status of each sample was determined according to the cut-off value of each mRNA; when the mRNA expression level in the sample was higher compared with the cut-off value, the status of this sample was defined as 1; otherwise, the status of this sample was defined as 0. Subsequently, a sample RS model was constructed using a linear combination of mRNA expression levels weighted by a regression coefficient (β) (obtained using the Cox regression model); thus, a prognostic index of each sample (i.e., RS) was calculated as follows: RS = Σ [β (mRNAn) × status (mRNAn)]. The RS of each sample in the training set was calculated, and the training samples were divided into prognostic high- (samples with an RS value ≥ median RS value) and low-risk groups (samples with an RS value < the median RS value) according to the median RS value. The prognostic difference between the high- and low-risk groups was evaluated using Kaplan-Meier curves and the significance was calculated using the log-rank test, and the effectiveness of the RS model was estimated by the area under the receiver-operating characteristic curve. In the validation set, the expression level of each mRNA was converted to the same probability distributions as that of the training dataset using Z score transformation, and the prognosis of the samples in the validation set was evaluated using the RS model.

Construction of a nomogram to study the association between independent prognostic factors and survival prognosis

In order to further investigate the association between clinical data and survival prognosis, the mRNAs identified to be significantly associated with prognosis were used to develop a nomogram using RMS package version 5.1–2 in R 3.4.0 (https://cran.r-project.org/web/packages/rms/index.html) (30). The scoring criteria were established according to the regression coefficients of all independent variables. Each value of an independent variable was assigned a score, and a total score was calculated for each sample. The probability of the outcome for each sample was calculated from the total score using a transforming function. The probability evaluation was performed using the nomogram method (31), and the nomogram was used to assess the association between the clinical factors and prognosis.

Screening and pathway analysis of mRNAs associated with prognostic risk

The training dataset samples were divided into high- and low-risk groups according to the RS. The differences between the groups in the mRNA expression matrix of the samples were analyzed using the edgeR package with false discovery rate <0.05 and |log2(fold-change)|>1 applied as thresholds to define significant differences. Pathway enrichment analysis was performed on the mRNAs with significant differential expression between the high- and low-risk groups using the GSEA (32). P<0.05 was selected as the threshold for identification of Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways significantly enriched in the DEG set (33).

Results

Identification of mRNAs with significant differential expression between groups with different prognosis

According to the source tissue, the 348 samples in the training set were divided into 311 STAD and 37 control samples, and the differences in the mRNA expression levels between the two groups were analyzed using the edgeR package. A total of 1,597 significantly differentially expressed mRNAs were identified (Fig. 2A). The results of hierarchical clustering analysis revealed a significant difference in the gene expression patterns between the tumor and control samples (Fig. 2B).

Screening for prognosis-associated genes and clinical factors

In the training set, univariate Cox regression analysis was performed on the 1,597 significantly differentially expressed mRNAs, 92 of which were identified to be significantly associated with survival prognosis (P<0.05; data not shown). In addition, the clinical information associated with the samples in the training set was subjected to univariate and multivariate Cox regression analyses; the results demonstrated that age, radiation therapy and recurrence were significantly associated with prognosis and were significant independent prognostic factors (Table II).

Table II.

Prognostic analysis of clinical factors in the training dataset.

Table II.

Prognostic analysis of clinical factors in the training dataset.

UnivariateMultivariate


VariablesHR (95% CI)P-valueHR (95% CI)P-value
Age, years1.023 (1.005–1.041)0.013a2.293 (1.334~3.943)0.003a
Sex, male/female1.476 (0.989–2.201)0.055
Subtype, MSI-H/MSI-L/MSS/-1.191 (0.940–1.508)0.146
Reflux, yes/no/-0.718 (0.370–1.393)0.325
Antireflux treatment, yes/no/-0.899 (0.499–1.619)0.723
H. pylori infection, yes/no/-0.519 (0.222–1.212)0.123
Radiation therapy, yes/no/-0.467 (0.279–0.780)0.003a0.459 (0.223–0.948)0.035a
Pathological_M, M0/M1/-2.480 (1.292–4.760)0.005a1.917 (0.666–5.518)0.228
Pathological_N, N0/N1/N2/N31.282 (1.091–1.506)0.002a1.062 (0.765–1.476)0.719
Pathological_T, T1/T2/T3/T4/-1.340 (1.067–1.684)0.012a1.339 (0.895–2.005)0.156
Pathological stage, I/II/III/IV/-1.565 (1.248–1.964) <0.001a1.208 (0.675–2.159)0.525
Grade, 1/2/3/41.351 (0.947–1.927)0.020a1.513 (0.869–2.633)0.143
Recurrence, yes/no/-2.261 (1.407–3.635) <0.001a1.334 (1.047–3.943)0.001a

a P<0.05. Tumors were staged according to the 7th edition TNM staging system (55). ‘−’, data not available; HR, hazard ratio; CI, confidence interval; MSI-H, microsatellite instability-high; MSI-L, microsatellite instability-low; MSS, microsatellite stability.

Establishment and evaluation of the RS model
Selection of an optimal mRNA combination

The expression matrix of the 92 mRNAs significantly associated with prognosis in the training set was selected as the input, and mRNA combinations were screened for optimal results using the LASSO Cox regression model in the penalized package. In the cyclic execution of the 1,000 CVL algorithm, the maximum CVL value (−730.883) was obtained with λ=21.178 (Fig. 3A and B), and 10 mRNAs were selected based on this parameter: Hemoglobin β (HBB), chromosome 4 open reading frame 48 (C4orf48), mannosidase endo-α-like (MANEAL), C-X-C motif chemokine ligand 3 (CXCL3), tripartite motif-containing 31 (TRIM31), transmembrane protein 200A (TMEM200A), serpin family E member 1 (SERPINE1), coagulation factor V (F5), NADPH oxidase organizer 1 (NOXO1) and Dickkopf WNT signaling pathway inhibitor 1 (DKK1 (Table III; Fig. 3C).

Table III.

Optimized mRNA combination.

Table III.

Optimized mRNA combination.

GenecoefHR (95%CI)P-value
HBB0.2701.115 (1.012–1.230)0.028
C4orf480.1911.206 (1.043–1.394)0.011
MANEAL−0.7260.756 (0.639–0.895)0.001
CXCL3−0.2400.891 (0.807–0.984)0.023
TRIM31−0.5670.869 (0.788–0.959)0.005
TMEM200A0.0221.234 (1.026–1.484)0.025
SERPINE10.6631.236 (1.093–1.397)0.001
F50.4291.147 (1.025–1.282)0.016
NOXO1−0.2880.782 (0.669–0.914)0.002
DKK10.7291.129 (1.045–1.220)0.002

[i] Coef, coefficient; HR, hazard ratio; CI, confidence interval; HBB, hemoglobin β; C4orf48, chromosome 4 open reading frame 48; MANEAL, mannosidase endo-α-like; CXCL3, C-X-C motif chemokine ligand 3; TRIM31, tripartite motif-containing 31; TMEM200A, transmembrane protein 200A; SERPINE1, serpin family E member 1; F5, coagulation factor V; NOXO1, NADPH oxidase organizer 1; DKK1, Dickkopf WNT signaling pathway inhibitor 1.

Determination of the mRNA expression cutoff values

For each mRNA included in the optimal mRNA combination, the expression level cut-off value was selected using the X-Tile Software with the Monte-Carlo P<0.05. As presented in Fig. 4, the cut-off values of HBB, C4orf48, DKK1, F5, NOXO1, SERPINE1, CXCL3, TMEM200A, MANEAL and TRIM31 were 6.4, 4.4, 1.6, 3.0, 1.2, 3.8, 5.8, 0.9, 1.4 and 0.9, respectively. Improved survival time was observed in patients with high expression levels of HBB, C4orf48, DKK1, F5, SERPINE1 and TMEM200A compared with patients with high expression levels of these genes, as well as in patients with low expression levels of NOXO1, CXCL3, MANEAL and TRIM31 compared with patients with low expression levels of these genes (Fig. 4).

Figure 4.

X-Tile analysis results on (A) HBB, (B) C4orf48 and (C) DKK1. Cyan and gray bars indicate the number of samples exhibiting high- and low-expression levels, respectively. The number at the junction of two colors represents the cutoff value of for high- and low-expression samples. In the right panel, cyan and gray Kaplan-Meier curves represent mRNAs in the high- and low-expression groups, respectively. HBB, hemoglobin β; C4orf48, chromosome 4 open reading frame 48; MANEAL, mannosidase endo-α-like; CXCL3, C-X-C motif chemokine ligand 3; TRIM31, tripartite motif-containing 31; TMEM200A, transmembrane protein 200A; SERPINE1, serpin family E member 1; F5, coagulation factor V; NOXO1, NADPH oxidase organizer 1; DKK1, Dickkopf WNT signaling pathway inhibitor 1; AUC, area under the curve. X-Tile analysis results on (D) F5, (E) NOXO1 and (F) SERPINE1. Cyan and gray bars indicate the number of samples exhibiting high- and low-expression levels, respectively. The number at the junction of two colors represents the cutoff value of for high- and low-expression samples. In the right panel, cyan and gray Kaplan-Meier curves represent mRNAs in the high- and low-expression groups, respectively. HBB, hemoglobin β; C4orf48, chromosome 4 open reading frame 48; MANEAL, mannosidase endo-α-like; CXCL3, C-X-C motif chemokine ligand 3; TRIM31, tripartite motif-containing 31; TMEM200A, transmembrane protein 200A; SERPINE1, serpin family E member 1; F5, coagulation factor V; NOXO1, NADPH oxidase organizer 1; DKK1, sDickkopf WNT signaling pathway inhibitor 1; AUC, area under the curve. X-Tile analysis results on (G) CXCL3, (H) TMEM200A (I) MANEAL. Cyan and gray bars indicate the number of samples exhibiting high- and low-expression levels, respectively. The number at the junction of two colors represents the cutoff value of for high- and low-expression samples. In the right panel, cyan and gray Kaplan-Meier curves represent mRNAs in the high- and low-expression groups, respectively. HBB, hemoglobin β; C4orf48, chromosome 4 open reading frame 48; MANEAL, mannosidase endo-α-like; CXCL3, C-X-C motif chemokine ligand 3; TRIM31, tripartite motif-containing 31; TMEM200A, transmembrane protein 200A; SERPINE1, serpin family E member 1; F5, coagulation factor V; NOXO1, NADPH oxidase organizer 1; DKK1, Dickkopf WNT signaling pathway inhibitor 1; AUC, area under the curve. X-Tile analysis results on (J) CXCL3. Cyan and gray bars indicate the number of samples exhibiting high- and low-expression levels, respectively. The number at the junction of two colors represents the cutoff value of for high- and low-expression samples. In the right panel, cyan and gray Kaplan-Meier curves represent mRNAs in the high- and low-expression groups, respectively. HBB, hemoglobin β; C4orf48, chromosome 4 open reading frame 48; MANEAL, mannosidase endo-α-like; CXCL3, C-X-C motif chemokine ligand 3; TRIM31, tripartite motif-containing 31; TMEM200A, transmembrane protein 200A; SERPINE1, serpin family E member 1; F5, coagulation factor V; NOXO1, NADPH oxidase organizer 1; DKK1, Dickkopf WNT signaling pathway inhibitor 1; AUC, area under the curve.

The RS prediction model was established as follows: RS = 0.27 × status (HBB) + 0.191 × status (C4orf48) + (−0.726) × status (MANEAL) + (−0.240) × status (CXCL3) + (−0.567) × status (TRIM31) + 0.022 × status (TMEM200A) + 0.663 × status (SERPINE1) + 0.429 × status (F5) + (−0.288) × status (NOXO1) + 0.729 × Status (DKK1). The RS prediction model was used to evaluate and verify the risk prediction effect for samples in the training and validation datasets (Fig. 5). According to the median RS value, high- and low-risk stratified analyses of the samples were performed. As exhibited in Table IV, five clinical factors were revealed to be associated with overall survival time according to univariable analysis, including age, pathological T, pathological stage, radiation therapy and recurrence in the low-risk group (P<0.05). Further multivariable Cox regression analysis revealed that radiation therapy was an independent clinical factor associated with STAD prognosis (P=0.035). In the high-risk group, H. pylori infection, pathological T, pathological N, pathological M, pathological stage and tumor grade were significantly associated with overall survival time (P<0.05). Among these factors, pathological N was an independent clinical factor for STAD prognosis (P=0.011).

Figure 5.

Risk prediction effect of the RS model for samples in the (A) training and (B) validation datasets. (A-a) The Kaplan-Meier curves of overall survival in the training dataset. Green and red curves denote low- and high-risk samples, respectively. (A-b) The Kaplan-Meier curves of recurrence-free survival in the training set. Blue and purple curves represent low- and high-risk samples, respectively. (A-c) The ROC curve for the training set. Black and purple indicate ROC curves for overall and recurrence-free survival, respectively. (B-a) The Kaplan–Meier curves of overall survival in the validation set. Green and red curves represent low- and high-risk samples, respectively. (B-b) The Kaplan-Meier curves of recurrence-free survival in the validation set. Blue and purple curves represent low- and high-risk samples, respectively. (B-c) The ROC curve for the validation set. Black and purple indicate ROC curves for overall and recurrence-free survival, respectively. ROC, receiver operating characteristic; AUC, area under the curve. Risk prediction effect of the RS model for samples in the (A) training and (B) validation datasets. (A-a) The Kaplan-Meier curves of overall survival in the training dataset. Green and red curves denote low- and high-risk samples, respectively. (A-b) The Kaplan-Meier curves of recurrence-free survival in the training set. Blue and purple curves represent low- and high-risk samples, respectively. (A-c) The ROC curve for the training set. Black and purple indicate ROC curves for overall and recurrence-free survival, respectively. (B-a) The Kaplan-Meier curves of overall survival in the validation set. Green and red curves represent low- and high-risk samples, respectively. (B-b) The Kaplan-Meier curves of recurrence-free survival in the validation set. Blue and purple curves represent low- and high-risk samples, respectively. (B-c) The ROC curve for the validation set. Black and purple indicate ROC curves for overall and recurrence-free survival, respectively. ROC, receiver operating characteristic; AUC, area under the curve.

Table IV.

Stratified analysis of clinicopathological characteristics in the high- and low-risk groups.

Table IV.

Stratified analysis of clinicopathological characteristics in the high- and low-risk groups.

A, Low-risk group (n=155)

UnivariateMultivariate


VariablesHR (95% CI)P-valueHR (95% CI)P-value
Age, years1.060 (1.020–1.103)0.003a1.041 (0.99–1.096)0.118
Sex, male/female1.207 (0.613–2.375)0.586
Subtype, MSI-H/MSI-L/MSS/-0.938 (0.650–1.353)0.732
Reflux, yes/no/-1.240 (1.092–2.647)0.997
H. pylori infection, yes/no/-0.969 (0.286–3.287)0.960
Pathological T, T1-T2/T3-T4/-1.320 (0.865–2.016)0.198
Pathological N, N0-N1/N2-N3/-1.367 (1.014–1.843)0.040a1.022 (0.598–1.746)0.937
Pathological M, M0/M1/-1.148 (0.273–4.821)0.851
Pathological stage, I/II/III/IV/-1.804 (1.158–2.810)0.009a2.006 (0.872–4.612)0.101
Grade, 1/2/3/40.882 (0.476–1.637)0.691
Anti-reflux treatment, yes/no/-1.177 (0.443–3.130)0.743
Radiation therapy, yes/no/-0.198 (0.068–0.575)0.003a0.197 (0.044–0.889)0.035a
Recurrence, yes/no/-3.622 (1.466–8.948)0.005a1.911 (0.691–5.289)0.212

B, High-risk group (n=156)

Univariate Multivariate


VariablesHR (95% CI)P-valueHR (95% CI)P-value

Age, years1.012 (0.992–1.032)0.247
Sex, male/female1.334 (0.808–2.204)0.260
Subtype, MSI-H/MSI-L/MSS/-1.138 (0.818–1.583)0.444
Reflux, yes/no/-0.751 (0.378–1.492)0.413
H. pylori infection, yes/no/-0.166 (0.038–0.725)0.017a0.175 (0.039–0.777)0.171
Pathological T, T1-T2/T3-T4/-1.542 (1.157–2.054)0.003a1.689 (0.757–3.771)0.201
Pathological N, N0-N1/N2+N3/-1.307 (1.077–1.586)0.007a1.790 (1.145–2.796)0.011a
Pathological M, M0/M1/-3.965 (1.858–8.465) <0.001a5.620 (1.297–14.348)0.199
Pathological stage, I/II/III/IV/-1.643 (1.254–2.152) <0.001a0.963 (0.465–1.992)0.918
Grade, 1/2/3/41.552 (0.995–2.420)0.053
Anti-reflux treatment, yes/no/-0.490 (0.209–1.148)0.101
Radiation therapy, yes/no/-0.902 (0.501–1.624)0.730
Recurrence, yes/no/-1.342 (0.767–2.349)0.302

a P<0.05. ‘−’, data not available; HR, hazard ratio; CI, confidence interval; MSI-H, microsatellite instability-high; MSI-L, microsatellite instability-low; MSS, microsatellite stability. Tumors were staged according to the 7th edition TNM staging system (55).

Association between clinical factors and survival prognosis according to the nomogram analysis

In the training set, three clinical factors (age, radiation therapy and recurrence) were subjected to stratified analysis in order to study the differences between the low- and high-risk groups for each clinical factor (Fig. 6). As shown in Fig. 6, the low-risk group exhibited longer overall survival times than the high-risk group.

In order to further analyze the association between age, radiation treatment, recurrence, and risk score with survival prognosis, a nomogram analysis was performed. Cox regression analysis suggested these four clinical factors were all significantly associated with survival prognosis (P<0.05, Fig. 7A). Then, a nomogram was established to predict the survival time of the samples. According to the nomogram, the 3- and 5- year survival probability may be predicted by matching the clinical indicators to the ‘Total Points’ axis (Fig. 7B).

Pathway analysis of the mRNAs associated with prognosis

The training dataset was subdivided into high- and low-risk groups according to the RS, and the differences in the mRNA expression matrix of the samples between these groups were analyzed using the edgeR package (Fig. 8A). A total of 728 significant DEGs were identified, including 221 downregulated and 507 upregulated DEGs. The samples were sorted according to the cor value and were clustered using the top 100 DEGs (top 50 positive and negative cor values; Fig. 8B).

GSEA pathway enrichment annotation was performed on the DEGs significantly associated with risk factors, and a total of eight significant KEGG pathways were identified (Table V). The heatmap of the genes involved in each pathway and the association between gene expression levels and pathways are presented in Fig. 9.

Table V.

KEGG pathways significantly associated with risk grouping.

Table V.

KEGG pathways significantly associated with risk grouping.

NameESNESNOM P-value
KEGG_ADHERENS_JUNCTION0.6081.2880.017
KEGG_TGF_BETA_SIGNALING_PATHWAY0.6051.1670.030
KEGG_ECM_RECEPTOR_INTERACTION0.5671.0760.041
KEGG_WNT_SIGNALING_PATHWAY0.5241.0930.042
KEGG_JAK_STAT_SIGNALING_PATHWAY0.4291.0090.043
KEGG_MAPK_SIGNALING_PATHWAY0.4861.0500.045
KEGG_PATHWAYS_IN_CANCER0.3661.0150.047
KEGG_MTOR_SIGNALING_PATHWAY0.5891.0170.049

[i] KEGG, Kyoto Encyclopedia of Genes and Genomes; ES, enrichment score; NES, Normalized enrichment score; NOM P-value, nominal P-value.

Discussion

The aberrant expression levels of certain genes are significantly associated with the pathogenesis and prognosis of GC (34). In the present study, a large amount of mRNA expression profiling data and clinical information from patients with STAD documented in TCGA database were used to identify statistically significant DEGs between STAD and healthy tissues. A total of 92 mRNAs significantly associated with survival were obtained by univariate Cox regression analysis.

The risk assessment tools that are based on gene expression levels can identify high-risk populations in relation to a specific disease and may be useful in the subsequent clinical decision-making process for healthcare providers and patients (12). In order to construct a risk assessment tool for the assessment of prognosis in patients with STAD, an optimal mRNA combination was identified among the 92 significant DEGs comprising 10 mRNAs in the present study, which were selected as a prognostic gene signature to create the RS model. The effectiveness of this model was evaluated in the training and validation datasets, and the results suggested that this risk assessment tool was useful for identifying populations with a high risk of developing STAD. To the best of our knowledge, aside from the present study, there is only one published RS model constructed according to gene expression levels, which includes a 53-gene signature and a prognostic scoring system (20). There are two other risk assessment tools that have been developed for the Japanese population, and their risk factors include age, sex, the combination of an anti-H. pylori antibody and serum pepsinogen, HbA1c level, smoking status, family history of GC and consumption of high-salt food (12,18). The risk assessment tool developed in the present study is simple and inexpensive enough to be used both in normal clinical practice and for mass screening.

The results of the present study revealed an improved prognosis for patients with high expression levels of HBB, C4orf48, DKK1, F5, SERPINE1 and TMEM200A and low expression levels of NOXO1, CXCL3, MANEAL and TRIM31. Although there are limited relevant studies on F5 and TMEM200A, certain functions of the remaining genes have been investigated. HBB is a globin protein constituting the most common form of hemoglobin in adult humans, and abnormal expression of HBB can lead to blood diseases, such as hemoglobinopathy and hereditary nigremia (11,35). C4orf48 is a gene identified in the Wolf-Hirschhorn syndrome critical chromosomal region that encodes a putative neuropeptide and is important for development of the neocortex and cerebellar (36) and cell differentiation (37). DKK1 protein is a soluble inhibitor of WNT that serves important roles in skeletal development (38) and is associated with the presence of lytic bone lesions in patients with multiple myeloma (39). SERPINE1, also known as plasminogen activator inhibitor type 1, is a member of the serine protease inhibitor family and is the major physiological regulator of the urokinase-type plasminogen activator-dependent pericellular plasmin-generating cascade (40,41). SERPINE1 has also been reported to serve roles in in acute lymphoblastic leukemia (42) and keratinocyte migration (43).

With the exception of MANEAL, which may be involved in neurological disorders (44), the other three genes with high expression levels in the samples with good prognosis are associated with cancer, particularly NOXO1 and TRIM31 (4549). NOXO1 can be induced in tumor epithelial cells and serves an important role in tumorigenicity and the tumor-initiating property of GC cells (46). In addition, NOXO1 may affect colon epithelium homeostasis and prevent inflammation (45). Previous studies have demonstrated that CXCL3 may be a biomarker of breast and prostate cancer (47,49). TRIM31, which is a ring finger, B-box and coiled-coil protein upregulated in GC cells and a potential biomarker of GC, can inhibit cell proliferation (48), and its cellular level may be regulated by a number of mechanisms, including the ubiquitin-proteasome system (50). GSEA pathway enrichment annotation revealed that eight pathways were enriched in the DEGs significantly associated with the risk factors, such as the ‘adherens junction’, ‘TGF-β signaling pathway’, ‘Wnt signaling pathway’, ‘JAK-STAT signaling pathway’, ‘MAPK signaling pathway’, ‘mTOR signaling pathway’ and ‘pathways in cancer’, all of which serve substantial roles in human carcinogenesis (5154). The functions of these prognostic genes are different compared with those of the genes identified in a previous study, which were associated with the cell cycle, RNA/non-coding RNA processes, acetylation and extracellular-matrix organization (20).

Independent validation of two different datasets and previous studies indicate that the RS model developed in the present study may be effective. However, a limitation of the present study was that it was an extensive bioinformatics analysis based on published data; the results should be validated using in vitro or in vivo models. However, the results of the present study may help other investigators to conduct relevant research.

In conclusion, a risk assessment tool for assessing the prognosis of patients with STAD was developed and validated in the present study. The 10 identified prognostic mRNAs were associated with several cellular processes and signaling pathways, such as the ‘adherens junction’, TGF-β signaling pathway’, ‘Wnt signaling pathway’, ‘JAK-STAT signaling pathway’, ‘MAPK signaling pathway’, ‘mTOR signaling pathway’ and ‘pathways in cancer’, and may be recommended as promising prognostic biomarkers or a prognostic signature of STAD. The present risk assessment tool may help identify patients with a high risk of STAD, and the proposed prognostic mRNAs may help elucidate the pathogenesis of STAD.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

ZL designed the present study. EG and FT conducted the bioinformatics analysis. EG contributed to writing the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

CVL

cross-validation likelihood

DEG

differentially expressed gene

GC

gastric cancer

GEO

Gene Expression Omnibus

GSEA

Gene Set Enrichment Analysis

KEGG

Kyoto Encyclopedia of Genes and Genomes

RNA-seq

high-throughput RNA sequencing

RS

risk score

STAD

stomach adenocarcinoma

TCGA

The Cancer Genome Atlas

References

1 

Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D and Bray F: Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 136:E359–E386. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Rugge M, Fassan M and Graham DY: Epidemiology of Gastric Cancer. Springer International Publishing; New York, NY: pp. 23–34. 2015

3 

Sanei MH, Mirmosayyeb O, Chehrei A, Ansari J and Saberi E: 5-year survival in gastric adenocarcinoma with epithelial and stromal versican expression. Iran J Pathol. 14:26–32. 2019. View Article : Google Scholar : PubMed/NCBI

4 

He XK and Sun LM: The increasing trend in the incidence of gastric cancer in the young population, not only in young Hispanic men. Gastric Cancer. 20:10102017. View Article : Google Scholar : PubMed/NCBI

5 

Merchant SJ, Kim J, Choi AH, Sun V, Chao J and Nelson R: A rising trend in the incidence of advanced gastric cancer in young Hispanic men. Gastric Cancer. 20:226–234. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Li Z, Lei H, Luo M, Wang Y, Dong L, Ma Y, Liu C, Song W, Wang F, Zhang J, et al: DNA methylation downregulated mir-10b acts as a tumor suppressor in gastric cancer. Gastric Cancer. 18:43–54. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Ng EK, Chong WW, Jin H, Lam EK, Shin VY, Yu J, Poon TC, Ng SS and Sung JJ: Differential expression of microRNAs in plasma of patients with colorectal cancer: A potential marker for colorectal cancer screening. Gut. 58:1375–1381. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Shikata K, Doi Y, Yonemoto K, Arima H, Ninomiya T, Kubo M, Tanizaki Y, Matsumoto T, Iida M and Kiyohara Y: Population-based prospective study of the combined influence of cigarette smoking and Helicobacter pylori infection on gastric cancer incidence: The Hisayama Study. Am J Epidemiol. 168:1409–1415. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Yamagata H, Kiyohara Y, Aoyagi K, Kato I, Iwamoto H, Nakayama K, Shimizu H, Tanizaki Y, Arima H, Shinohara N, et al: Impact of Helicobacter pylori infection on gastric cancer incidence in a general Japanese population: The Hisayama study. Arch Intern Med. 160:1962–1968. 2000. View Article : Google Scholar : PubMed/NCBI

10 

Shikata K, Kiyohara Y, Kubo M, Yonemoto K, Ninomiya T, Shirota T, Tanizaki Y, Doi Y, Tanaka K, Oishi Y, et al: A prospective study of dietary salt intake and gastric cancer incidence in a defined Japanese population: The Hisayama study. Int J Cancer. 119:196–201. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Ueda T, Volinia S, Okumura H, Shimizu M, Taccioli C, Rossi S, Alder H, Liu CG, Oue N, Yasui W, et al: Relation between microRNA expression and progression and prognosis of gastric cancer: A microRNA expression analysis. Lancet Oncol. 11:136–146. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Iida M, Ikeda F, Hata J, Hirakawa Y, Ohara T, Mukai N, Yoshida D, Yonemoto K, Esaki M, Kitazono T, et al: Development and validation of a risk assessment tool for gastric cancer in a general Japanese population. Gastric Cancer. 21:383–390. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Pavlou M, Ambler G, Seaman SR, Guttmann O, Elliott P, King M and Omar RZ: How to develop a more accurate risk prediction model when there are few events. BMJ. 351:h38682015. View Article : Google Scholar : PubMed/NCBI

14 

Hussein AA, Ghani KR, Peabody J, Sarle R, Abaza R, Eun D, Hu J, Fumo M, Lane B, Montgomery JS, et al: Development and validation of an objective scoring tool for robot-assisted radical prostatectomy: Prostatectomy assessment and competency evaluation. J Urol. 197:1237–1244. 2017. View Article : Google Scholar : PubMed/NCBI

15 

Nimeri AA, Bautista J, Ibrahim M, Philip R, Al Shaban T, Maasher A and Altinoz A: Mandatory risk assessment reduces venous thromboembolism in bariatric surgery patients. Obes Surg. 28:541–547. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Aminian A, Andalib A, Khorgami Z, Cetin D, Burguera B, Bartholomew J, Brethauer SA and Schauer PR: Who should get extended thromboprophylaxis after bariatric surgery?: A risk assessment tool to guide indications for post-discharge pharmacoprophylaxis. Ann Surg. 265:143–150. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Kastrinos F, Uno H, Ukaegbu C, Alvero C, McFarland A, Yurgelun MB, Kulke MH, Schrag D, Meyerhardt JA, Fuchs CS, et al: Development and validation of the PREMM5 model for comprehensive risk assessment of lynch syndrome. J Clin Oncol. 35:2165–2172. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Charvat H, Sasazuki S, Inoue M, Iwasaki M, Sawada N, Shimazu T, Yamaji T and Tsugane S; JPHC Study Group, : Prediction of the 10-year probability of gastric cancer occurrence in the Japanese population: The JPHC study cohort II. Int J Cancer. 138:320–331. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Yuan SQ, Wu WJ, Qiu MZ, Wang ZX, Yang LP, Jin Y, Yun JP, Gao YH, Li YH, Zhou ZW, et al: Development and validation of a nomogram to predict the benefit of adjuvant radiotherapy for patients with resected gastric cancer. J Cancer. 8:3498–3505. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Wang P, Wang Y, Hang B, Zou X and Mao JH: A novel gene expression-based prognostic scoring system to predict survival in gastric cancer. Oncotarget. 7:55343–55351. 2016.PubMed/NCBI

21 

Tibshirani R: The lasso method for variable selection in the Cox model. Stat Med. 16:385–395. 1997. View Article : Google Scholar : PubMed/NCBI

22 

Cristescu R, Lee J, Nebozhyn M, Kim KM, Ting JC, Wong SS, Liu J, Yue YG, Wang J, Yu K, et al: Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat Med. 21:449–456. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Robinson MD, McCarthy DJ and Smyth GK: edgeR: A Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 26:139–140. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Wang L, Cao C, Ma Q, Zeng Q, Wang H, Cheng Z, Zhu G, Qi J, Ma H, Nian H and Wang Y: RNA-seq analyses of multiple meristems of soybean: Novel and alternative transcripts, evolutionary and functional implications. BMC Plant Biol. 14:1692014. View Article : Google Scholar : PubMed/NCBI

25 

Eisen MB, Spellman PT, Brown PO and Botstein D: Cluster analysis and display of genome-wide expression patterns. Proc Natl Acad Sci USA. 95:14863–14868. 1998. View Article : Google Scholar : PubMed/NCBI

26 

Therneau T: A Package for Survival Analysis in S. version 2.38. https://CRAN.R-project.org/package=survival2015

27 

Thernea u, Terry M, Grambsc h and Patricia M: Modeling Survival Data: Extending the Cox Model. Springer; New York, NY: 2000, View Article : Google Scholar

28 

Goeman JJ: L1 penalized estimation in the Cox proportional hazards model. Biom J. 52:70–84. 2010.PubMed/NCBI

29 

Bachoc F: Cross validation and maximum likelihood estimations of hyper-parameters of gaussian processes with model misspecification. Comput Stat Data Anal. 66:55–69. 2013. View Article : Google Scholar

30 

Camp RL, Dolled-Filhart M and Rimm DL: X-tile: A new bio-informatics tool for biomarker assessment and outcome-based cut-point optimization. Clin Cancer Res. 10:7252–7259. 2004. View Article : Google Scholar : PubMed/NCBI

31 

Eng KH, Schiller E and Morrell K: On representing the prognostic value of continuous gene expression biomarkers with the restricted mean survival curve. Oncotarget. 6:36308–36318. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Iasonos A, Schrag D, Raj GV and Panageas KS: How to build and interpret a nomogram for cancer prognosis. J Clin Oncol. 26:1364–1370. 2008. View Article : Google Scholar : PubMed/NCBI

33 

Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES and Mesirov JP: Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA. 102:15545–15550. 2005. View Article : Google Scholar : PubMed/NCBI

34 

Kanehisa M and Goto S: KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 28:27–30. 2000. View Article : Google Scholar : PubMed/NCBI

35 

Piel FB, Howes RE, Patil AP, Nyangiri OA, Gething PW, Bhatt S, Williams TN, Weatherall DJ and Hay SI: The distribution of haemoglobin C and its prevalence in newborns in Africa. Sci Rep. 3:16712013. View Article : Google Scholar : PubMed/NCBI

36 

Thom CS, Dickson CF, Gell DA and Weiss MJ: Hemoglobin variants: Biochemical properties and clinical correlates. Cold Spring Harb Perspect Med. 3:a0118582013. View Article : Google Scholar : PubMed/NCBI

37 

Endele S, Nelkenbrecher C, Bördlein A, Schlickum S and Winterpacht A: C4ORF48, a gene from the Wolf-Hirschhorn syndrome critical region, encodes a putative neuropeptide and is expressed during neocortex and cerebellar development. Neurogenetics. 12:155–163. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Deeb A: Diabetes mellitus secondary to acute pancreatitis in a child with Wolf-Hirschhorn syndrome. Case Rep Endocrinol. 2017:38924672017.PubMed/NCBI

39 

Grotewold L and Rüther U: The Wnt antagonist Dickkopf-1 is regulated by Bmp signaling and c-Jun and modulates programmed cell death. EMBO J. 21:966–975. 2002. View Article : Google Scholar : PubMed/NCBI

40 

Tian EM, Zhan F, Walker R, Rasmussen E, Ma Y, Barlogie B and Shaughnessy JD Jr: The role of the Wnt-signaling antagonist DKK1 in the development of osteolytic lesions in multiple myeloma. N Engl J Med. 349:2483–2494. 2003. View Article : Google Scholar : PubMed/NCBI

41 

Providence KM, White LA, Tang J, Gonclaves J, Staiano-Coico L and Higgins PJ: Epithelial monolayer wounding stimulates binding of USF-1 to an E-box motif in the plasminogen activator inhibitor type 1 gene. J Cell Sci. 115:3767–3777. 2002. View Article : Google Scholar : PubMed/NCBI

42 

Turchi L, Chassot AA, Rezzonico R, Yeow K, Loubat A, Ferrua B, Lenegrate G, Ortonne JP and Ponzio G: Dynamic characterization of the molecular events during in vitro epidermal wound healing. J Investig Dermatol. 119:56–63. 2002. View Article : Google Scholar : PubMed/NCBI

43 

French D, Hamilton LH, Mattano LA Jr, Sather HN, Devidas M, Nachman JB and Relling MV; Children's Oncology Group, : A PAI-1 (SERPINE1) polymorphism predicts osteonecrosis in children with acute lymphoblastic leukemia: A report from the Children's Oncology Group. Blood. 111:4496–4499. 2008. View Article : Google Scholar : PubMed/NCBI

44 

Providence KM, Higgins SP, Mullen A, Battista A, Samarakoon R, Higgins CE, Wilkins-Port CE and Higgins PJ: SERPINE1 (PAI-1) is deposited into keratinocyte migration ‘trails’ and required for optimal monolayer wound repair. Arch Dermatol Res. 300:303–310. 2008. View Article : Google Scholar : PubMed/NCBI

45 

Herebian D, Alhaddad B, Seibt A, Schwarzmayr T, Danhauser K, Klee D, Harmsen S, Meitinger T, Strom TM, Schulz A, et al: Coexisting variants in OSTM1 and MANEAL cause a complex neurodegenerative disorder with NBIA-like brain abnormalities. Eur J Hum Genet. 25:1092–1095. 2017. View Article : Google Scholar : PubMed/NCBI

46 

Moll F, Walter M, Rezende F, Helfinger V, Vasconez E, De Oliveira T, Greten FR, Olesch C, Weigert A, Radeke HH and Schröder K: NoxO1 controls proliferation of colon epithelial cells. Front Immunol. 9:9732018. View Article : Google Scholar : PubMed/NCBI

47 

Oshima H, Ishikawa T, Yoshida GJ, Naoi K, Maeda Y, Naka K, Ju X, Yamada Y, Minamoto T, Mukaida N, et al: TNF-α/TNFR1 signaling promotes gastric tumorigenesis through induction of Noxo1 and Gna14 in tumor cells. Oncogene. 33:3820–3829. 2014. View Article : Google Scholar : PubMed/NCBI

48 

Gui SL, Teng LC, Wang SQ, Liu S, Lin YL, Zhao XL, Liu L, Sui HY, Yang Y, Liang LC, et al: Overexpression of CXCL3 can enhance the oncogenic potential of prostate cancer. Int Urol Nephrol. 48:701–709. 2016. View Article : Google Scholar : PubMed/NCBI

49 

Sugiura T and Miyamoto K: Characterization of TRIM31, upregulated in gastric adenocarcinoma, as a novel RBCC protein. J Cell Biochem. 105:1081–1091. 2008. View Article : Google Scholar : PubMed/NCBI

50 

See AL, Chong PK, Lu SY and Lim YP: CXCL3 is a potential target for breast cancer metastasis. Curr Cancer Drug Targets. 14:294–309. 2014. View Article : Google Scholar : PubMed/NCBI

51 

Sugiura T: The cellular level of TRIM31, an RBCC protein overexpressed in gastric cancer, is regulated by multiple mechanisms including the ubiquitin-proteasome system. Cell Biol Int. 35:657–661. 2013. View Article : Google Scholar

52 

Zhan T, Rindtorff N and Boutros M: Wnt signaling in cancer. Oncogene. 36:1461–1473. 2017. View Article : Google Scholar : PubMed/NCBI

53 

Neuzillet C, Tijeras-Raballand A, Cohen R, Cros J, Faivre S, Raymond E and de Gramont A: Targeting the TGFβ pathway for cancer therapy. Pharmacol Ther. 147:22–31. 2015. View Article : Google Scholar : PubMed/NCBI

54 

Zou X and Blank M: Targeting p38 MAP kinase signaling in cancer through post-translational modifications. Cancer Lett. 384:19–26. 2017. View Article : Google Scholar : PubMed/NCBI

55 

Waddell T, Verheij M, Allum W, Cunningham D, Cervantes A and Arnold D; European Society for Medical Oncology (ESMO); European Society of Surgical Oncology (ESSO); European Society of Radiotherapy Oncology (ESTRO), : Gastric cancer: ESMO-ESSO-ESTRO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 24 (Suppl 6):vi57–vi63. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2020
Volume 19 Issue 2

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Guan E, Tian F and Liu Z: A novel risk score model for stomach adenocarcinoma based on the expression levels of 10 genes. Oncol Lett 19: 1351-1367, 2020
APA
Guan, E., Tian, F., & Liu, Z. (2020). A novel risk score model for stomach adenocarcinoma based on the expression levels of 10 genes. Oncology Letters, 19, 1351-1367. https://doi.org/10.3892/ol.2019.11190
MLA
Guan, E., Tian, F., Liu, Z."A novel risk score model for stomach adenocarcinoma based on the expression levels of 10 genes". Oncology Letters 19.2 (2020): 1351-1367.
Chicago
Guan, E., Tian, F., Liu, Z."A novel risk score model for stomach adenocarcinoma based on the expression levels of 10 genes". Oncology Letters 19, no. 2 (2020): 1351-1367. https://doi.org/10.3892/ol.2019.11190