Open Access

An 8‑gene signature predicts the prognosis of cervical cancer following radiotherapy

  • Authors:
    • Fei Xie
    • Dan Dong
    • Na Du
    • Liang Guo
    • Weihua Ni
    • Hongyan Yuan
    • Nannan Zhang
    • Jiang Jie
    • Guomu Liu
    • Guixiang Tai
  • View Affiliations

  • Published online on: July 29, 2019     https://doi.org/10.3892/mmr.2019.10535
  • Pages: 2990-3002
  • Copyright: © Xie 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

Gene expression and DNA methylation levels affect the outcomes of patients with cancer. The present study aimed to establish a multigene risk model for predicting the outcomes of patients with cervical cancer (CerC) treated with or without radiotherapy. RNA sequencing training data with matched DNA methylation profiles were downloaded from The Cancer Genome Atlas database. Patients were divided into radiotherapy and non‑radiotherapy groups according to the treatment strategy. Differently expressed and methylated genes between the two groups were identified, and 8 prognostic genes were identified using Cox regression analysis. The optimized risk model based on the 8‑gene signature was defined using the Cox's proportional hazards model. Kaplan‑Meier survival analysis indicated that patients with higher risk scores exhibited poorer survival compared with patients with lower risk scores (log‑rank test, P=3.22x10‑7). Validation using the GSE44001 gene set demonstrated that patients in the high‑risk group exhibited a shorter survival time comprared with the low‑risk group (log‑rank test, P=3.01x10‑3). The area under the receiver operating characteristic curve values for the training and validation sets were 0.951 and 0.929, respectively. Cox regression analyses indicated that recurrence and risk status were risk factors for poor outcomes in patients with CerC treated with or without radiotherapy. The present study defined that the 8‑gene signature was an independent risk factor for the prognosis of patients with CerC. The 8‑gene prognostic model had predictive power for CerC prognosis.

Introduction

Cervical cancer (CerC) is a leading cause of cancer-associated mortality in women worldwide (1,2). Surgery in combination with chemotherapy and radiotherapy is the most common strategy for CerC treatment. Radiotherapy significantly improves CerC patient prognosis (3). However, the overall survival of patients with CerC diagnosed at advanced stages remains poor, with the 5-year survival rate ≤50%, despite advanced surgical protocols and diagnostic methods (4).

The general prognosis criteria of the International Federation of Gynecology and Obstetrics (FIGO) classification do not include all prognostic factors, including histologic subtypes and lymph node metastasis, which are effective for the prediction of CerC prognosis (5). Molecular markers and clinical parameters are crucial for the prediction of clinical outcomes and deciding treatment strategies (2,5). In addition, the identification of biomarkers associated with radiotherapy response is of great importance for understanding the molecular mechanisms of CerC and developing novel strategies.

Radiotherapy significantly benefits patients with CerC (3). The methylation status in the promoters of a number of genes is associated with patient outcomes after radiotherapy (68). For example, Dunn et al (6) demonstrated that the O6-methyxlguanine-DNA-methyltransferase (MGMT) promoter methylation level was positively associated with the progression-free survival and overall survival of patients with glioblastomas treated with temozolomide and radiotherapy (6). Huang et al (7) indicated that the combined Ras association domain family member (RASSF) 1A/RASSF2A methylation level was negatively correlated with the disease-free survival (DFS) of radiotherapy-treated squamous cell carcinoma. Widschwendter et al (9) revealed that the methylated myoblast determination protein 1 (MYOD1) in CerC was associated with poor DFS (9).

An increasing number of studies have indicated the prognostic power of gene signatures for diease prognosis, metastasis and recurrence. Okayama et al (10) identified a 4 gene signature with prediction power for stage I lung cancer prognosis (10); Cheng et al (11) described an 8-gene classifier with predictive power for locoregional recurrence of breast cancer in patients post-mastectomy (11). Therefore, the predictive power of multigene sigatures for disease development may be of great clinical interest. A 12-gene classifier has been used for the clinical diagnosis of low and high metastasis of in uveal melanoma (12,13). In addition, the DNA methylation level is a significant factor in disease development (68). However, to the best of our knowledge, there have been few studies investigating gene methylation signatures for prognosis in radiotherapy-treated patients with CerC.

The present study was designed to explore a novel risk model for predicting outcome of patients with CerC by analyzing RNA sequencing (RNA-seq) data in combination with matched DNA methylation profiles from The Cancer Genome Atlas (TCGA) database. A multigene risk model that predicted the outcomes of patients with CerC treated with or without radiotherapy was identified.

Materials and methods

TCGA and Gene Expression Omnibus (GEO) dataset

Training data were downloaded from TCGA database (https://gdc-portal.nci.nih.gov/) in June 2018. A total of 307 mRNA-seq profiles (Illumina Hiseq2000) and 312 DNA methylation profiles (Illumina Infinium Human Methylation 450 BeadChip) were downloaded. Paired mRNA-seq and methylation data were included in the present study. Clinical features including age, pathologic stage and grade, and survival rate of patients with CerC were extracted and used for subsequent analysis.

Validation dataset GSE44001 (GPL14951 Illumina HumanHT-12 WG-DASL V4.0 R2 expression beadchip) (14) was downloaded from the National Center of Biotechnology Information GEO database (http://www.ncbi.nlm.nih.gov/geo/). The GSE44001 dataset consists of 300 patients with primary early CerC (FIGO stage I–II). Prognostic data were available for the training and validation sets. The study design presented in Fig. 1.

Identification of differentially expressed and methylated genes

Samples from the TCGA training set were assigned into two groups according to radiotherapy treatment (with and without radiotherapy). Differentially expressed genes (DEGs) and differentially methylated genes (DMGs) between the two groups were identified using Linear Models for Microarray Data (Limma) package (version 3.34.7) in R (https://bioconductor.org/packages/release/bioc/html/limma.html). A false discovery rate (FDR) <0.05 and |log2 fold change (FC) | >0.263 (>1.2 FC) were set as the cutoffs. DEGs with differential methylation were selected.

Correlation analysis between gene expression and methylation level

Pearson's correlation between gene expression and the methylation level of DEGs was calculated using Cor. Test function (https://stat.ethz.ch/R-manual/R-devel/library/stats/html/cor.test.html) in R. DEGs with significantly correlated expression and methylation level (P<0.05) were included as candidate genes for subsequent analysis.

Selection of prognostic DEGs

Univariate Cox regression analysis in survival package of R (version 2.41.3; http://CRAN.R-project.org/package=survival) (15) was used to screen the DEGs and DMGs associated with the prognosis of patients with CerC. P<0.05, determined by a Kaplan-Meier log-rank test, was defined as the significant cutoff value.

Definition and validation of prognostic risk model

Cox's proportional hazards (Cox-PH) model based on the L1-penalized least absolute shrinkage and selection operator regression algorithm in the penalized package (version 0.9.50) was used for optimizing a prediction model with a linear gene signature (16). The optimized parameter ‘lamba’ was obtained by 1,000 rounds of cross-validated likelihood (cvl) circular calculation. The risk score of each sample was defined as the linear combination of prognostic gene expression level and Cox-PH regression coefficient: Risk score=∑coefgene × Expression (Methylation) gene. Patients were assigned into high-risk and low-risk groups according to the median value of risk score. The overall survival difference between the two groups was evaluated using Kaplan-Meier and log-rank methods in survival package of R (version 2.41.3; http://CRAN.R-project.org/package=survival). The GSE44001 dataset was used to validate the performance and predictive power of the prognostic risk model. The area under the time-independent receiver operating characteristic curve (AUC) was used for evaluation (2).

Selection and stratification analyses of potential clinical prognostic factors

The independent prognostic risk factors among clinical variables in TCGA patients were selected using univariate and multivariate Cox regression analysis in survival package of R (https://CRAN.R-project.org/package=survival). P<0.05, determined by a Kaplan-Meier log-rank test, was set as the significant cutoff value. Stratification analysis was performed for patients with and without radiotherapy, with a significant threshold of P<0.05, as determined by a log-rank test.

Bioinformatic analysis of prognostic DEGs

Patients within the training set were assigned into high-risk (samples with higher risk scores than the median) and low-risk (samples with lower risk scores than the median) groups according to the computed risk scores. DEGs between the two groups (FDR <0.05 and |logFC| >0.263) were identified using Limma package in R. Hierarchical clustering analysis of DEGs was analyzed using the Pheatmap package (version 1.0.8; http://CRAN.R-project.org/package=pheatmap) in R (17,18). Gene Set Enrichment Analysis (GSEA) (19,20) was performed to identify the Kyoto Encyclopedia of Genes and Genomes (KEGG) (21) pathways significantly (P<0.05) associated with DEGs between the two groups.

Statistical analysis

Continuous clinical variables, including age and overall survival, are presented as the mean ± standard deviation (SD), and differences between groups were analyzed using Student's t-test. Differences in categorical variables, including mortality and pathological characteristics, between two groups were analyzed using Fisher's exact test. Univariate Cox regression analysis was employed for the identification of independent prognostic genes, and a two-step Cox regression analysis was used to identify independent prognostic factors among clinical variables. In the stratified analysis, prognostic differences between the high-risk and low-risk patients stratification analysis were analyzed using Kaplan-Meier survival analysis. All analyses were performed in R (version 3.4.1; http://www.r-project.org/), and P<0.05 was considered to indicate a statistically significantly difference.

Results

Baseline characteristics of patients with CerC

A total of 238 patients with CerC with paired mRNA-seq and DNA methylation profiles from TCGA were used in the present study. Table I describes the baseline characteristics of the included 238 patients. A total of 64 and 174 patients were assigned into non-radiotherapy and radiotherapy groups, respectively. Significant differences in age (P=3.12×10−2), pathologic N stage (P=2.15×10−3), pathologic T stage (P=1.90×10−5), pathologic stage (P=9.84×10−5), new tumor incidence (recurrence; P=2.20×10−16) and therapy strategy (P=3.16×10−10) were observed between patients with and without radiotherapy. There was no difference in overall survival and survival rate between the two groups (Table I).

Table I.

Baseline characteristics of The Cancer Genome Atlas patients with cervical cancer treated with or without radiotherapy.

Table I.

Baseline characteristics of The Cancer Genome Atlas patients with cervical cancer treated with or without radiotherapy.

Clinical characteristicsWithout radiotherapy (N=64)With radiotherapy (N=174)P-value
Age, years, mean ± SD45.09±11.4949.01±14.19 3.12×10−2a
Pathologic M (M0/M1/NA)29/1/3457/9/108 1.65×10−1b
Pathologic N (N0/N1/NA)45/7/1258/35/81 2.15×10−3b
Pathologic T (T1/T2/T3/T4/NA)47/9/0/4/462/46/17/5/44 1.902×10−5b
Pathologic stage (I/II/III/IV/NA)49/8/2/4/178/47/29/16/4 9.842×10−5b
Pathologic grade (1/2/3/4/NA)6/27/26/0/510/79/66/1/18 6.92×10−1b
Smoking (reformed/current/never/NA)7/12/38/733/43/86/12 2.06×10−2b
New tumor (yes/no/-)51/12/131/142/1 2.20×10−16b
Targeted molecular therapy (yes/no/NA)5/25/34127/37/10 3.164×10−10b
Death (dead/alive)15/4951/123 4.17×10−1b
Overall survival months, mean ± SD35.11±43.0338.75±38.92 5.54×10−1a

a Student's t test.

b Fisher's exact test. NA, not available; SD, standard deviation.

Identification of DEGs and DMGs in patients with CerC

There were 1,488 DEGs and 2,888 DMGs identified between the two groups (Fig. 2); the majority of the DEGs (62.28%, 1,016/1,488) were upregulated and the majority of the DMGs (58.14%, 1,679/2,888) were hypomethylated by radiotherapy, compared with the non-radiotherapy group (Fig. 2A and B). There were 203 overlapping genes, with an overall negative correlation between average expression and methylation levels (Cor=−0.4045; P=2.16×10−9; Fig. 2C). Pearson's correlation analysis identified 107 genes (including 83 up- and 24 downregulated genes, Table SI) with negatively correlated expression and methylation levels.

Identification of prognostic genes

Using univariate Cox regression analysis in the survival package of R, a total of 25 prognostic DEGs and 21 prognostic DMGs were identified from the aforementioned 107 genes (Table SII), including 8 overlapped genes, which were identified to be candidate genes associated with the prognosis of patients with CerC (Fig. 3A). The optimal 8-gene matrix was obtained using Cox-PH model (max ‘lambda’=5.8598, max cvl=−367.5751; Fig. 3B). The Cox-PH regression coefficients are indicated in Fig. 3C and Table II. Accordingly, 238 patients in the training set were stratified into high-expression (n=119) and low-expression (n=119) groups, according to the median expression value of each gene.

Table II.

Cox's proportional hazards regression coefficients of the 8 signature genes.

Table II.

Cox's proportional hazards regression coefficients of the 8 signature genes.

GeneCorrelation coefficientHR (95% CI)P-value
CCDC136−0.62410.917 (0.758–0.991) 2.71×10−2
ABCG2−1.20370.847 (0.699–0.925) 4.76×10−2
CYP26A1−1.01680.889 (0.786–0.998) 6.00×10−3
TNNI3−1.10940.881 (0.799–0.971) 4.87×10−2
SYT13   0.69231.076 (1.008–1.168) 4.52×10−2
FOXC2   0.67091.066 (1.056–1.189) 2.12×10−3
CXCL5   0.70991.075 (1.006–1.160) 2.48×10−3
TMEM233   1.56231.234 (1.007–1.526) 2.61×10−4

[i] HR, hazard ratio; CI, confidence interval; CCDC136, coiled-coil domain containing 136 gene; ABCG2, ATP binding cassette subfamily G member 2 gene; CY26A1, cytochrome P450 26A1 gene; TNNI3, cardiac troponin I gene; SYT13, synaptotagmin XIII gene; FOXC2, Forkhead 1 gene; CXCL5, epithelial neutrophil-activating peptide-78 gene; TMEM233, transmembrane protein 233 gene.

Subsequent Kaplan-Meier survival analyses demonstrated that the expression of coiled-coil domain containing 136 gene (CCDC136), ATP binding cassette subfamily G member 2 gene (ABCG2), cardiac troponin I gene (TNNI3) and Cytochrome P450 26A1 gene (CYP26A1) were positively associated with survival of patients with CerC, whereas the expression of Synaptotagmin XIII gene (SYT13), Forkhead 1 gene (FOXC2), epithelial neutrophil-activating peptide-78 gene (CXCL5) and transmembrane protein 233 gene (TMEM233) were negatively correlated (log-rank test; P<0.05; Fig. 4A; Table II). For methylation levels, analysis indicated the hypermethylation of CCDC136, ABCG2, TNNI3, and CYP26A1 genes, and the hypomethylation of SYT13, FOXC2, CXCL5, and TMEM233 genes was associated with the poor survival of patients with CerC (P<0.05, log-rank test; Fig. 4B). These data demonstrated that the expression and methylation levels of these 8 genes were potential independent risk factors for prognosis in patients with CerC.

Establishment and evaluation of the risk model

The mRNA prognostic model based on the combination of Cox-PH regression coefficients and gene expression levels was established as: Risk score=(−0.6241) × ExpCCDC136+ (−1.2037) × ExpABCG2 + (−1.0168) × ExpCYP26A1+ (−1.1094) × ExpTNNI3+ (0.6923) × ExpSYT13 + (0.6709) × ExpFOXC2+ (0.7099) × ExpCXCL5 + (1.5623) × ExpTMEM233. According to the median risk score, 238 patients with CerC in the training set were assigned into high-risk and low-risk groups. Kaplan-Meier survival analysis indicated that patients with low-risk scores exhibited longer overall survival compared with patients with high-risk scores (P=3.22×10−7, log-rank test; Fig. 5A). The AUC was 0.951 (Fig. 5B). Analysis of the validation set GSE44001 demonstrated that patients with CerC with high-risk scores exhibited significantly shorter overall survival times compared with patients with low-risk scores (P=3.01×10−3, log-rank test; Fig. 5C), and the AUC was 0.929 (Fig. 5D). These results demonstrated that the 8-gene signature had performance and predictive power for outcomes of patients with CerC.

Prognostic value of clinical variables

A two-step Cox regression analyses (univariate and multivariate) were used to define the potential prognostic values of clinical variables, including age, pathologic stage and grade, smoking, radiotherapy, recurrence and risk status, in patients from the TCGA data. Table III demonstrates that 3 independent risk factors, including pathologic stage [hazard ratio (HR)=2.386; 95% confidence interval (CI), 1.097–5.192; P=0.0284), new tumor (recurrence; HR=7.333; 95% CI, 1.833–12.235; P=3.21×10−9) and risk status (HR=1.359; 95% CI, 1.702–8.905; P=1.28×10−3) were of prognostic value for the outcomes of patients with CerC. Kaplan-Meier survival analysis determined the prognostic potential of pathologic stage and tumor recurrence. As presented in Fig. 6, there was a significantly shorter overall survival time in patients with advanced (III–IV) pathological stages (P=3.21×10−5, log-rank test; Fig. 6A) and recurrence (P=9.22×10−15, log-rank test; Fig. 6B), compared with patients in early stage disease without recurrence.

Table III.

Cox regression analyses for the prognostic value of clinical variables.

Table III.

Cox regression analyses for the prognostic value of clinical variables.

Univariate analysisMultivariate analysis


VariablesHR (95% CI)P-valueHR (95% CI)P-value
Age (≤45/>45)1.014 (0.996–1.032)0.138
Pathologic M (M0/M1)4.101 (1.356–12.41)0.0677
Pathologic N (N0/N1)2.923 (1.440–5.932) 1.86×10−31.535 (0.711–3.315) 2.76×10−1
Pathologic T (T1/T2/T3/T4)1.986 (1.473–2.678) 2.69×10−61.900 (0.986–3.662) 5.51×10−2
Pathologic stage (I/II/III/IV)1.594 (1.277–1.989) 2.20×10−52.386 (1.097–5.192) 2.84×10−2
Pathologic grade (1/2/3/4)0.943 (0.612–1.452) 7.89×10−1
Smoking (reformed/current/never)0.984 (0.719–1.346) 9.18×10−1
New tumor (yes/no)5.637 (3.446–9.22) 9.22×10−157.33 (1.833–12.235) 3.22×10−9
Targeted molecular therapy (yes/no)0.953 (0.547–1.659) 8.64×10−1
Risk status (high/low)3.736 (2.177–6.411) 3.22×10−71.359 (1.702–8.905) 1.28×10−3

[i] HR, hazard ratio; CI, confidence interval.

Stratification analysis for risk factors associated with radiotherapy

To additionally confirm the risk factors associated with radiotherapy, stratified analysis for patients with radiotherapy and without radiotherapy was performed. A two-step Cox regression analyses indicated that pathologic N stage (HR=4.247; 95% CI, 1.3651–6.216; P=1.25×10−2), pathologic stage (HR=2.275; 95% CI, 1.052–3.868; P=4.53×10−2), recurrence (HR=3.841; 95% CI, 1.332–5.122; P=2.27×10−5) and risk status (HR=5.110; 95% CI, 1.578–6.547; P=6.51×10−3) were risk factors for radiotherapy-treated patients, whereas recurrence (HR=4.665; 95% CI, 2.367–9.463; P=1.58×10−3) and risk status (HR=7.546; 95% CI, 1.177–8.364; P=3.30×10−2) were risk factors for patients treated without radiotherapy (Table IV). These results demonstrated that recurrence and 8-gene signature risk status were independent risk factors for predicting the prognosis of patients with CerC.

Table IV.

Stratification analysis for risk factors associated with radiotherapy in patients from The Cancer Genome Atlas training set.

Table IV.

Stratification analysis for risk factors associated with radiotherapy in patients from The Cancer Genome Atlas training set.

Univariate analysisMultivariate analysis


VariablesHR95% CIP-valueHR95% CIP-value
With radiotherapy (n=174)
Age (≤45/>45)1.0060.986–1.026 5.48×10−1
Pathologic M (M0/M1)5.4571.436–10.73 5.13×10−2
Pathologic N (N0/N1)4.6511.769–12.23 6.31×10−44.2471.3651–6.216 1.25×10−2
Pathologic T (T1/T2/T3/T4)1.6521.135–2.407 7.65×10−31.5640.7868–3.107 2.02×10−1
Pathologic stage (I/II/III/IV)2.3201.327–4.056 2.36×10−32.2751.052–3.868 4.53×10−2
Pathologic grade (1/2/3/4)0.8780.540–1.428 6.01×10−1
Smoking (reformed/current/never)0.9980.705–1.415 9.95×10−1
New tumor (yes/no)5.1912.986–9.026 7.89×10−113.8411.332–5.122 2.27×10−5
Targeted molecular therapy (yes/no)1.060.539–2.081 .66×10−1
Risk status (high/low)3.1981.762–5.804 5.51×10−55.111.578–6.547 6.51×10−3
Without radiotherapy (n=64)
Age (≤45/>45)1.0511.004–1.1 3.13×10−21.0310.977–1.089 2.67×10−1
Pathologic M (M0/M1)
Pathologic N (N0/N1)1.3480.287–2.337 7.05×10−1
Pathologic T (T1/T2/T3/T4)3.6381.999–6.623 2.29×10−91.1870.122–1.598 8.83×10−1
Pathologic stage (I/II/III/IV)2.4621.281–3.148 4.53×10−52.6551.243–4.007 4.05×10−1
Pathologic grade (1/2/3/4)1.1920.438–3.245 7.31×10−1
Smoking (reformed/current/never)0.8630.395–1.882 7.10×10−1
New tumor (yes/no)7.8022.53–14.06 2.52×10−54.6652.367–9.463 1.58×10−3
Targeted molecular therapy (yes/no)1.4080.998–2.181 2.77×10−1
Risk status (high/low)6.7621.804–10.35 1.40×10−37.5461.177–8.364 3.30×10−2

[i] HR, hazard ratio; CI, confidence interval. Risk status was predicated based on the 8-gene risk model.

Identification of DEGs and KEGG pathways associated with risk status of patients with CerC

To define the gene profiles between patients with high and low risk status, 490 DEGs were identified (Table SIII) in the high-risk group, compared with the low-risk group (Fig. 7A), including 313 upregulated DEGs (63.88%, including CXCL5, SYT13, FOXC2, ITGB3 and TMEM233) and 177 downregulated DEGs (36.18%, including CYP26A1 and TNNI3) in the high-risk group. Fig. 7B demonstrates the markedly altered expression profiles of these DEGs in patients with low and high risk scores. GSEA KEGG pathway analysis indicated that these genes (including CYP26A1 and CXCL5) were associated with pathways including ‘ECM Receptor Interaction’, ‘Retinol Metabolism’, ‘Focal Adhesion’, ‘Hedgehog Signaling Pathway’, ‘NOD-like Receptor Signaling Pathway’ and ‘Chemokine Signaling Pathway’ (Table V).

Table V.

Gene Set Enrichment Analysis of the KEGG pathways associated with differentially expressed genes between patients with cervical cancer with high- and low-risk scores.

Table V.

Gene Set Enrichment Analysis of the KEGG pathways associated with differentially expressed genes between patients with cervical cancer with high- and low-risk scores.

KEGG termESNESNOM P-valueGene
ECM receptor interaction0.76951.2517 1.88×10−2LAMA1, COL11A1, ITGB3, IBSP
Retinol Metabolism−0.8045−1.2455 2.05×10−2ADH7, CYP26A1, CYP26C1, UGT2A1
Focal adhesion0.69631.2640 2.14×10−2LAMA1, COL11A1, ITGB3
Hedgehog signaling pathway−0.7371−1.2183 2.34×10−2WNT3A, BMP7
NOD-like receptor signaling pathway0.74131.1071 3.98×10−2CXCL2, IL6, IL1B
Chemokine signaling pathway0.50710.9989 4.72×10−2CXCL2, CXCL6, ADCY1, CXCL3, CXCL5

[i] KEGG, Kyoto Encyclopedia of Genes and Genomes; ECM, extracellular matrix; ES, enrichment score; NES, normalized enrichment score; NOM, nominal.

Discussion

Identification of molecular biomarkers associated with radiotherapy may aid in devising strategies for improving radiotherapy response (22). In the present study, a large-scale analysis of RNA-seq from TCGA CerC samples, in combination with matched DNA methylation profiles, was performed, and an 8-gene risk model was identified (CCDC136, ABCG2, CYP26A1, TNNI3, CXCL5, SYT13 FOXC2, ITGB3, and TMEM233) to predict the risk status of patients with CerC. This 8-gene signature was defined to be an independent prognostic factor, with predictive power for prognosis of patients with CerC. Among these 8 genes, 4 hypermethylated genes (CCDC136, ABCG2, CYP26A1 and TNNI3) were positively associated the overall survival of patients with CerC, and 4 hypomethylated genes (SYT13, FOXC2, CXCL5 and TMEM233) were negatively associated the overall survival of patients with CerC.

The 4 hypermethylated genes (CCDC136, ABCG2, CYP26A1 and TNNI3) had previously been identified to be dysregulated in various human cancer tissues (2326). Among these, TNNI3 is an angiogenesis inhibitor responsible for the inhibition of endothelial cell tube formation (27,28). Kern et al (28) suggested that metastasis was decreased in a mouse model of pancreatic cancer in response to troponin I treatment, compared with control mice. Downregulated troponin I inhibits cancer cell proliferation, as it is required for tumor growth (29). CCDC136, also known as nasopharyngeal carcinoma-associated gene 6, is located at chromosome 7q31-32. It is commonly deleted in a number of types of malignant human cancer, and has been recognized to function as a putative tumor suppressor in gastric tumor and nasopharyngeal carcinoma (23,30). Wei et al (30) suggested that CCDC136 negatively regulated the Wnt/β-catenin signaling pathway in zebrafish embryos. Wnt/β-catenin signaling is oncogenic and confers cancer cell proliferation, drug resistance and metastasis in various types of human cancer, including ovarian cancer and CerC (3134) (Fig. 8). In the present study, it was identified that CCDC136 and TNNI3 were upregulated in the radiotherapy group, compared with the non-radiotherapy group. This may be associated with the decreased angiogenesis and downregulated Wnt/β-catenin signaling in patients treated with radiotherapy, which in turn is associated with the lower recurrence and improved prognosis observed in the radiotherapy group.

ABCG2 encodes an multidrug transporter protein, breast cancer resistance protein (BCRP), which contributes to drug resistance in cancer cell lines and tumors (35,36). It has been reported that ABCG2 is downstream of Wnt/β-catenin signaling and is responsible for chemoresistance (37). Downregulated BCRP/ABCG2 is common in tumor tissues, including CerC, which may function in tumorigenesis by promoting the accumulation of genotoxins and nitric oxide (24,38,39). In addition, ABCG2 promoter methylation has been described in multiple myeloma tissues (40). The demethylation of ABCG2 increases its expression and enhances multidrug resistance in cancer cells (40,41). CYP26A1 is an oncogenic protein in breast cancer, cervical squamous neoplasia, ovarian cancer, and head and neck cancer (25,26,42). CYP26A1 is a metabolizing enzyme for retinoic acids (RAs) (25). RAs induce the differentiation of various types of stem cells (43), and the RA receptor γ (RARγ) is associated with the Akt/NF-κB and Wnt/β-catenin signaling pathways in tumorigenesis (44). Yasuhara et al (45) suggested that RARγ enhances and inhibits Wnt/β-catenin signaling in RA-free and RA-treated conditions, respectively. Demethylation and hypermethylation of CYP26A1 had been demonstrated in the CYP26A1-positive T47D cell line, which exhibits low rates of metastasis, and the CYP26A1-negative T47D cell line, which exhibits high rates of metastasis, respectively (46). It has been suggested that increased methylation levels in the CYP26A1 promoter is associated with poor survival in patients with prostate cancer (47). In the present study, the expression levels of ABCG2 and CYP26A1 were downregulated and upregulated, respectively, in patients in the radiotherapy group compared with the non-radiotherapy group. These two genes were identified to be positively associated with the prognosis of patients with CerC, and their hypermethylation was correlated with poor survival. In addition, it was also observed that CYP26A1 was associated with the ‘Retinol Metabolism’ GSEA KEGG pathway, which was associated with RA metabolism in cancer cells (48). These results suggested the complex roles of these genes in response to radiotherapy, and their potential prognostic value.

Among the 4 hypomethylated genes (SYT13, FOXC2, CXCL5 and TMEM233), SYT13, FOXC2 and CXCL5 have been demonstrated to be associated with tumorigenesis. FOXC2 is a downstream target of the Akt/NF-κB signaling pathway and is critical for tumor metastasis (49). The inhibition of FOXC2 results in the suppression of tumor metastasis and chemoresistance in lung cancer cells, nasopharyngeal carcinomas and CerC cells (4951). Synaptotagmins are a family of Ca2+ sensors that function in promoting membrane fusion (52,53). Overexpression of synaptotagmin has previously been described in human cancer (5457). Kanda et al (58) demonstrated that SYT13 was upregulated in gastric cancer and was associated with metastatic status. CXCL5 is a CXC-type chemokine, and is involved in angiogenesis and associated with poor prognosis in cancer patients (5962). In addition, CXCL5 expression activated the Akt/NF-κB and Wnt/β-catenin signaling pathways (6365). FOXC2 and CXCL5 were upregulated in the patients treated with radiotherapy compared with patients without radiotherapy, and their expression was associated with poor survival in patients with CerC. These demonstrated the potential prognostic value of SYT13, FOXC2 and CXCL5 for predicting patients with high risk status or poor outcomes.

In conclusion, a significant difference in survival was observed between the patients with CerC with high- and low-risk scores according to the 8-gene signature. The AUC and survival analysis in the training and validation set revealed the performance and predictive power of the 8-gene signature risk model for predicting survival of patients with CerC. Cox regression analysis indicated that the 8-gene signature was an independent risk factor for the prognosis of patients with CerC. Validation with more and larger clinical cohorts may additionally verify the potential prognostic value of the 8-gene signature in patients with CerC.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

The present study was supported by grants from the Double Tenth Engineering of Major Research Project of Jilin Provincial Science and Technology Department (grant no. 20140201012YY) and the Major Development Programs for New Drugs of the Chinese Academy of Sciences during the 12th Five-Year Plan Period (grant no. 2011ZX09102-001-36).

Availability of data and materials

The results published here are in part based upon data generated by the TCGA Research Network (https://www.cancer.gov/tcga). The Gene Expression Omnibus dataset GSE44001 is available at http://www.ncbi.nlm.nih.gov/geo/. All data generated during this study are included in this published article.

Authors' contributions

FX, DD and GT were responsible for the conception and design of the research. ND, LG, WN, HY, NZ, JJ and GL acquired and analyzed the data. FX and DD drafted the manuscript. LG, WN, HY, NZ, JJ and GL revised important intellectual content. All authors agreed with the final revision.

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.

References

1 

Cancer Genome Atlas Research Network; Albert Einstein College of Medicine; Analytical Biological Services; Barretos Cancer Hospital; Baylor College of Medicine; Beckman Research Institute of City of Hope; Buck Institute for Research on Aging; Canada's Michael Smith Genome Sciences Centre; Harvard Medical School, . Helen F, et al: Integrated genomic and molecular characterization of cervical cancer. Nature. 543:378–384. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Huang K, Sun H, Li X, Hu T, Yang R, Wang S, Jia Y, Chen Z, Tang F, Shen J, et al: Prognostic risk model development and prospective validation among patients with cervical cancer stage IB2 to IIB submitted to neoadjuvant chemotherapy. Sci Rep. 6:275682016. View Article : Google Scholar : PubMed/NCBI

3 

Jadon R, Pembroke CA, Hanna CL, Palaniappan N, Evans M, Cleves AE and Staffurth J: A systematic review of organ motion and image-guided strategies in external beam radiotherapy for cervical cancer. Clin Oncol (R Coll Radiol). 26:185–196. 2014. View Article : Google Scholar : PubMed/NCBI

4 

White A, Joseph D, Rim SH, Johnson CJ, Coleman MP and Allemani C: Colon cancer survival in the United States by race and stage (2001–2009): Findings from the CONCORD-2 study. Cancer. 123 (Suppl 24):S5014–S5036. 2017. View Article : Google Scholar

5 

Obrzut B, Kusy M, Semczuk A, Obrzut M and Kluska J: Prediction of 5-year overall survival in cervical cancer patients treated with radical hysterectomy using computational intelligence methods. BMC Cancer. 17:8402017. View Article : Google Scholar : PubMed/NCBI

6 

Dunn J, Baborie A, Alam F, Joyce K, Moxham M, Sibson R, Crooks D, Husband D, Shenoy A, Brodbelt A, et al: Extent of MGMT promoter methylation correlates with outcome in glioblastomas given temozolomide and radiotherapy. Br J Cancer. 101:124–131. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Huang KH, Huang SF, Chen IH, Liao CT, Wang HM and Hsieh LL: Methylation of RASSF1A, RASSF2A, and HIN-1 is associated with poor outcome after radiotherapy, but not surgery, in oral squamous cell carcinoma. Clin Cancer Res. 15:4174–4180. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Miousse IR, Kutanzi KR and Koturbash I: Effects of ionizing radiation on DNA methylation: From experimental biology to clinical applications. Int J Radiat Biol. 93:457–469. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Widschwendter A, Müller HH, Fiegl H, Ivarsson L, Wiedemair A, Müller-Holzner E, Goebel G, Marth C and Widschwendter M: DNA methylation in serum and tumors of cervical cancer patients. Clin Cancer Res. 10:565–571. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Okayama H, Schetter AJ, Ishigame T, Robles AI, Kohno T, Yokota J, Takenoshita S and Harris CC: The expression of four genes as a prognostic classifier for stage I lung adenocarcinoma in 12 independent cohorts. Cancer Epidemiol Biomarkers Prev. 23:2884–2894. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Cheng SH, Horng CF, Huang TT, Huang ES, Tsou MH, Shi LS, Yu BL, Chen CM and Huang AT: An eighteen-gene classifier predicts locoregional recurrence in post-mastectomy breast cancer patients. EBioMedicine. 5:74–81. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Harbour JW: A prognostic test to predict the risk of metastasis in uveal melanoma based on a 15-gene expression profile. Methods Mol Biol. 1102:427–440. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Field MG, Decatur CL, Kurtenbach S, Gezgin G, van der Velden PA, Jager MJ, Kozak KN and Harbour JW: PRAME as an independent biomarker for metastasis in Uveal melanoma. Clin Cancer Res. 22:1234–1242. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Lee YY, Kim TJ, Kim JY, Choi CH, Do IG, Song SY, Sohn I, Jung SH, Bae DS, Lee JW and Kim BG: Genetic profiling to predict recurrence of early cervical cancer. Gynecol Oncol. 131:650–654. 2013. View Article : Google Scholar : PubMed/NCBI

15 

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

16 

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

17 

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

18 

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

19 

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

20 

Mootha VK, Lindgren CM, Eriksson KF, Subramanian A, Sihag S, Lehar J, Puigserver P, Carlsson E, Ridderstråle M, Laurila E, et al: PGC-1alpha-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat Genet. 34:267–273. 2003. View Article : Google Scholar : PubMed/NCBI

21 

Kanehisa M, Furumichi M, Tanabe M, Sato Y and Morishima K: KEGG: New perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res. 45:D353–D361. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Liu FF, Shi W, Done SJ, Miller N, Pintilie M, Voduc D, Nielsen TO, Nofech-Mozes S, Chang MC, Whelan TJ, et al: Identification of a Low-Risk luminal a breast cancer cohort that may not benefit from breast radiotherapy. J Clin Oncol. 33:2035–2040. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Zhang XM, Sheng SR, Wang XY, Bin LH, Wang JR and Li GY: Expression of tumor related gene NAG6 in gastric cancer and restriction fragment length polymorphism analysis. World J Gastroenterol. 10:1361–1364. 2004. View Article : Google Scholar : PubMed/NCBI

24 

Gupta N, Martin PM, Miyauchi S, Ananth S, Herdman AV, Martindale RG, Podolsky R and Ganapathy V: Down-regulation of BCRP/ABCG2 in colorectal and cervical cancer. Biochem Biophys Res Commun. 343:571–577. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Osanai M and Lee GH: Increased expression of the retinoic acid-metabolizing enzyme CYP26A1 during the progression of cervical squamous neoplasia and head and neck cancer. BMC Res Notes. 7:6972014. View Article : Google Scholar : PubMed/NCBI

26 

Downie D, Mcfadyen MC, Rooney PH, Cruickshank ME, Parkin DE, Miller ID, Telfer C, Melvin WT and Murray GI: Profiling cytochrome P450 expression in ovarian cancer: Identification of prognostic markers. Clin Cancer Res. 11:7369–7375. 2005. View Article : Google Scholar : PubMed/NCBI

27 

Wang D, Li T, Cui H and Zhang Y: Analysis of the indicating value of cardiac troponin I, tumor necrosis factor-α, interleukin-18, Mir-1 and Mir-146b for viral myocarditis among Children. Cell Physiol Biochem. 40:1325–1333. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Kern BE, Balcom JH, Antoniu BA, Warshaw AL and Fernández-del Castillo C: Troponin I peptide (Glu94-Leu123), a cartilage-derived angiogenesis inhibitor: In vitro and in vivo effects on human endothelial cells and on pancreatic cancer. J Gastrointest Surg. 7:961–969. 2003. View Article : Google Scholar : PubMed/NCBI

29 

Casas-Tintó S, Maraver A, Serrano M and Ferrús A: Troponin-I enhances and is required for oncogenic overgrowth. Oncotarget. 7:52631–52642. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Wei S, Shang H, Cao Y and Wang Q: The coiled-coil domain containing protein Ccdc136b antagonizes maternal Wnt/β-catenin activity during zebrafish dorsoventral axial patterning. J Genet Genomics. 43:431–438. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Kopczynski J, Kowalik A, Chłopek M, Wang ZF, Góźdź S, Lasota J and Miettinen M: Oncogenic activation of the Wnt/β-catenin signaling pathway in signet ring stromal cell tumor of the ovary. Appl Immunohistochem Mol Morphol. 24:e28–e33. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Nagaraj AB, Joseph P, Kovalenko O, Singh S, Armstrong A, Redline R, Resnick K, Zanotti K, Waggoner S and DiFeo A: Critical role of Wnt/β-catenin signaling in driving epithelial ovarian cancer platinum resistance. Oncotarget. 6:23720–23734. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Emons G, Spitzner M, Reineke S, Auslander N, Kramer F, Rave-Fraenk M, Gaedcke J, Ghadimi M, Ried T and Grade M: Abstract 4760: Wnt/β-catenin signaling mediates resistance of colorectal cancer cell lines to chemoradiotherapy. Cancer Res. 77:4760. 2017.

34 

Lan K, Zhao Y, Fan Y, Ma B, Yang S, Liu Q, Linghu H and Wang H: Sulfiredoxin may promote cervical cancer metastasis via Wnt/β-catenin signaling pathway. Int J Mol Sci. 18(pii): E9172017.PubMed/NCBI

35 

Sarkadi B, Ozvegy-Laczka C, Német K and Váradi A: ABCG2-a transporter for all seasons. FEBS Lett. 567:116–120. 2004. View Article : Google Scholar : PubMed/NCBI

36 

Elkind NB, Szentpétery Z, Apáti A, Ozvegy-Laczka C, Várady G, Ujhelly O, Szabó K, Homolya L, Váradi A, Buday L, et al: Multidrug transporter ABCG2 prevents tumor cell death induced by the epidermal growth factor receptor inhibitor Iressa (ZD1839, Gefitinib). Cancer Res. 65:1770–1777. 2005. View Article : Google Scholar : PubMed/NCBI

37 

Chau WK, Ip CK, Mak AS, Lai HC and Wong AS: c-Kit mediates chemoresistance and tumor-initiating capacity of ovarian cancer cells through activation of Wnt/β-catenin-ATP-binding cassette G2 signaling. Oncogene. 32:2767–2781. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Liu HG, Pan YF, You J, Wang OC, Huang KT and Zhang XH: Expression of ABCG2 and its significance in colorectal cancer. Asian Pac J Cancer Prev. 11:845–848. 2010.PubMed/NCBI

39 

Sari FM, Yanar HT and Ozhan G: Investigation of the functional single-nucleotide polymorphisms in the BCRP transporter and susceptibility to colorectal cancer. Biomed Rep. 3:105–109. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Turner JG, Gump JL, Zhang C, Cook JM, Marchion D, Hazlehurst L, Munster P, Schell MJ, Dalton WS and Sullivan DM: ABCG2 expression, function, and promoter methylation in human multiple myeloma. Blood. 108:3881–3889. 2006. View Article : Google Scholar : PubMed/NCBI

41 

Bram EE, Stark M, Raz S and Assaraf YG: Chemotherapeutic drug-induced ABCG2 promoter demethylation as a novel mechanism of acquired multidrug resistance 1 2. Neoplasia. 11:1359–1370. 2009. View Article : Google Scholar : PubMed/NCBI

42 

Chang CL, Hong E, Lao-Sirieix P and Fitzgerald RC: A novel role for the retinoic acid-catabolizing enzyme CYP26A1 in Barrett's associated adenocarcinoma. Oncogene. 27:2951–2960. 2008. View Article : Google Scholar : PubMed/NCBI

43 

Tang XH and Gudas LJ: Retinoids, retinoic acid receptors, and cancer. Annu Rev Pathol. 6:345–364. 2011. View Article : Google Scholar : PubMed/NCBI

44 

Huang GL, Luo Q, Rui G, Zhang W, Zhang QY, Chen QX and Shen DY: Oncogenic activity of retinoic acid receptor γ is exhibited through activation of the Akt/NF-κB and Wnt/β-catenin pathways in cholangiocarcinoma. Mol Cell Biol. 33:3416–3425. 2013. View Article : Google Scholar : PubMed/NCBI

45 

Yasuhara R, Yuasa T, Williams JA, Byers SW, Shah S, Pacifici M, Iwamoto M and Enomoto-Iwamoto M: Wnt/beta-catenin and retinoic acid receptor signaling pathways interact to regulate chondrocyte function and matrix turnover. J Biol Chem. 285:317–327. 2010. View Article : Google Scholar : PubMed/NCBI

46 

Francesca C, Stefano R, Gaia B, Ren M and Nicoletta S: Derangement of a factor upstream of RARalpha triggers the repression of a pleiotropic epigenetic network. PLoS One. 4:e43052009. View Article : Google Scholar : PubMed/NCBI

47 

Liu LY: Association of tissue promoter methylation levels of APC, RASSF1A, CYP26A1 and TBX15 with prostate cancer progression (unpublished PhD thesis)University of Toronto (Canada); 2012

48 

García-Mariscal A, Peyrollier K, Basse A, Pedersen E, Rühl R, van Hengel J and Brakebusch C: RhoA controls retinoid signaling by ROCK dependent regulation of retinol metabolism. Small GTPases. 9:433–444. 2018. View Article : Google Scholar : PubMed/NCBI

49 

Yu YH, Chen HA, Chen PS, Cheng YJ, Hsu WH, Chang YW, Chen YH, Jan Y, Hsiao M, Chang TY, et al: MiR-520h-mediated FOXC2 regulation is critical for inhibition of lung cancer progression by resveratrol. Oncogene. 32:431–443. 2013. View Article : Google Scholar : PubMed/NCBI

50 

Zheng CH, Quan Y, Li YY, Deng WG, Shao WJ and Fu Y: Expression of transcription factor FOXC2 in cervical cancer and effects of silencing on cervical cancer cell proliferation. Asian Pac J Cancer Prev. 15:1589–1595. 2014. View Article : Google Scholar : PubMed/NCBI

51 

Zhou Z, Zhang L, Xie B, Wang X, Yang X, Ding N, Zhang J, Liu Q, Tan G, Feng D and Sun LQ: FOXC2 promotes chemoresistance in nasopharyngeal carcinomas via induction of epithelial mesenchymal transition. Cancer Lett. 363:137–145. 2015. View Article : Google Scholar : PubMed/NCBI

52 

Martens S, Kozlov MM and Mcmahon HT: How synaptotagmin promotes membrane fusion. Science. 316:1205–1208. 2007. View Article : Google Scholar : PubMed/NCBI

53 

Geppert M, Goda Y, Hammer RE, Li C, Rosahl TW, Stevens CF and Südhof TC: Synaptotagmin I: A major Ca2+ sensor for transmitter release at a central synapse. Cell. 79:717–727. 1994. View Article : Google Scholar : PubMed/NCBI

54 

Jahn JE and Coleman WB: Phenotypic normalization of GN6TF rat liver tumor cells results from WT1 expression following transfection of human SYT13-containing BACs. FASEB J. 20:A10912006.

55 

Kanda M, Shimizu D, Tanaka H, Tanaka C, Kobayashi D, Hayashi M, Iwata N, Niwa Y, Yamada S, Fujii T, et al: Significance of SYT8 For the detection, prediction, and treatment of peritoneal metastasis from gastric cancer. Ann Surg. 267:495–503. 2016. View Article : Google Scholar

56 

Sung HY, Han J, Ju W and Ahn JH: Synaptotagmin-like protein 2 gene promotes the metastatic potential in ovarian cancer. Oncol Rep. 36:535–541. 2016. View Article : Google Scholar : PubMed/NCBI

57 

Jin H, Xu G, Zhang Q, Pang Q and Fang M: Synaptotagmin-7 is overexpressed in hepatocellular carcinoma and regulates hepatocellular carcinoma cell proliferation via Chk1-p53 signaling. Onco Targets Ther. 10:4283–4293. 2017. View Article : Google Scholar : PubMed/NCBI

58 

Kanda M, Shimizu D, Tanaka H, Tanaka C, Kobayashi D, Hayashi M, Takami H, Niwa Y, Yamada S, Fujii T, et al: Synaptotagmin XIII expression and peritoneal metastasis in gastric cancer. Br J Surg. 105:1349–1358. 2018. View Article : Google Scholar : PubMed/NCBI

59 

Li A, King J, Moro A, Sugi MD, Dawson DW, Kaplan J, Li G, Lu X, Strieter RM, Burdick M, et al: Overexpression of CXCL5 is associated with poor survival in patients with pancreatic cancer. Am J Pathol. 178:1340–1349. 2011. View Article : Google Scholar : PubMed/NCBI

60 

Park JY, Park KH, Bang S, Kim MH, Lee JE, Gang J, Koh SS and Song SY: CXCL5 overexpression is associated with late stage gastric cancer. J Cancer Res Clin Oncol. 133:835–840. 2007. View Article : Google Scholar : PubMed/NCBI

61 

Kawamura M, Toiyama Y, Tanaka K, Saigusa S, Okugawa Y, Hiro J, Uchida K, Mohri Y, Inoue Y and Kusunoki M: CXCL5, a promoter of cell proliferation, migration and invasion, is a novel serum prognostic marker in patients with colorectal cancer. Eur J Cancer. 48:2244–2251. 2012. View Article : Google Scholar : PubMed/NCBI

62 

Begley L, Kasina S, Mehra R, Adsule S, Admon AJ, Lonigro RJ, Chinnaiyan AM and Macoska JA: CXCL5 promotes prostate cancer progression. Neoplasia. 10:244–254. 2008. View Article : Google Scholar : PubMed/NCBI

63 

Wang C, Li A, Yang S, Qiao R, Zhu X and Zhang J: CXCL5 promotes mitomycin C resistance in non-muscle invasive bladder cancer by activating EMT and NF-κB pathway. Biochem Biophys Res Commun. 498:862–868. 2018. View Article : Google Scholar : PubMed/NCBI

64 

Guan Z, Li C, Fan J, He D and Li L: Androgen receptor (AR) signaling promotes RCC progression via increased endothelial cell proliferation and recruitment by modulating AKT→NF-κB→CXCL5 signaling. Sci Rep. 6:370852016. View Article : Google Scholar : PubMed/NCBI

65 

Zhao J, Ou B, Han D, Wang P, Zong Y, Zhu C, Liu D, Zheng M, Sun J, Feng H and Lu A: Tumor-derived CXCL5 promotes human colorectal cancer metastasis through activation of the ERK/Elk-1/Snail and AKT/GSK3β/β-catenin pathways. Mol Cancer. 16:702017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2019
Volume 20 Issue 4

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Xie F, Dong D, Du N, Guo L, Ni W, Yuan H, Zhang N, Jie J, Liu G, Tai G, Tai G, et al: An 8‑gene signature predicts the prognosis of cervical cancer following radiotherapy. Mol Med Rep 20: 2990-3002, 2019
APA
Xie, F., Dong, D., Du, N., Guo, L., Ni, W., Yuan, H. ... Tai, G. (2019). An 8‑gene signature predicts the prognosis of cervical cancer following radiotherapy. Molecular Medicine Reports, 20, 2990-3002. https://doi.org/10.3892/mmr.2019.10535
MLA
Xie, F., Dong, D., Du, N., Guo, L., Ni, W., Yuan, H., Zhang, N., Jie, J., Liu, G., Tai, G."An 8‑gene signature predicts the prognosis of cervical cancer following radiotherapy". Molecular Medicine Reports 20.4 (2019): 2990-3002.
Chicago
Xie, F., Dong, D., Du, N., Guo, L., Ni, W., Yuan, H., Zhang, N., Jie, J., Liu, G., Tai, G."An 8‑gene signature predicts the prognosis of cervical cancer following radiotherapy". Molecular Medicine Reports 20, no. 4 (2019): 2990-3002. https://doi.org/10.3892/mmr.2019.10535