Open Access

Prognostic and predictive value of immune/stromal-related gene biomarkers in renal cell carcinoma

  • Authors:
    • Sen Wang
    • Xiangguang Zheng
    • Xinglu Chen
    • Xiaojun Shi
    • Sansan Chen
  • View Affiliations

  • Published online on: April 24, 2020     https://doi.org/10.3892/ol.2020.11574
  • Pages: 308-316
  • Copyright: © Wang 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

Immune/stromal-associated genes may be promising biomarkers for cancer diagnosis and the determination of clinical cancer treatment options. The aim of the present study was to identify prognostic stromal/immune-associated genes in renal cell carcinoma (RCC). RCC gene expression data (885 cases) were obtained from The Cancer Genome Atlas database. Immune/stromal scores were calculated by using the ESTIMATE package in R. Immune/stromal scores were significantly associated with Tumor-Node-Metastasis stage, clinical stage and overall survival rate (P<0.05). There were 419 differentially expressed genes (DEGs) based on immune scores and 738 DEGs based on stromal scores. Among these DEGs, 406 DEGs based on stromal scores and 252 DEGs based on immune scores were significantly associated with overall survival rate (P<0.05). The biological functions of these DEGs were primarily enriched in the ‘immune response’ and ‘regulation of cell migration and proliferation’. These DEGs were observed in a protein-protein interaction network. A LASSO Cox regression model was used to build a prognostic 6 gene-based classifier, including the IL21R, ATP6V1C2, GBP1, P2RY10, GBP4 and TNNC2 genes [area under the curve (AUC) =0.776]. The predictive model which combined this classifier with clinical prognostic factors had a high accuracy in predicting patient survival in RCC (combined AUC =0.899). Taken together, these results demonstrated that there are significant associations between immune/stromal scores and clinicopathological staging. A set of tumor microenvironment-associated genes that have powerful prognostic value in patients with RCC were identified in the present study.

Introduction

Renal cell carcinoma (RCC) is one of the most common urological tumors worldwide, and its incidence and mortality have been rising throughout the world (1). Traditional chemotherapeutic drugs cannot easily kill kidney cancer cells (2) and the clinical outcomes of patients with RCC can vary greatly (3). At present, the prognosis of patients with RCC is primarily determined by pathological staging (4). Although various studies have been investigated RCC, the clinical prognosis of patients with metastatic RCC remains poor (5). Therefore, there is a need to identify novel and more effective and accurate prognosis biomarkers for patients with RCC. At present, gene expression profiling has been increasingly used for the diagnosis of RCC (6). The genomic and molecular characteristics of tumors, such as RCC and bladder cancer, have been investigated in recent years (7). Molecular biomarkers should be considered in addition to standard clinicopathologic criteria to guide clinical treatment options.

Tumor-associated genes are important contributors to tumorigenesis and progression of cancer (8,9). In addition, the tumor microenvironment affects gene expression levels in tumor tissues, thus affecting clinical outcomes (10,11). In the tumor microenvironment, immune and stromal cells are the two main types of non-tumor components and are associated with the prognosis of tumors (12,13). Gene expression levels data from tumor databases can be analyzed using algorithms to predict cellular components in tumor tissues (14,15). For example, Yoshihara et al. designed an algorithm called ESTIMATE (16), which uses expression data to estimate stromal and immune cells in malignant tumor tissue. The algorithm can predict the infiltration of immune and stromal cells in the tumor microenvironment by analyzing specific gene expression profiles and calculating immune and stromal scores. At present, the value of immune/stromal scores in RCC has not been elucidated in detail. Novel useful biomarkers are required to estimate the prognosis and diagnosis of RCC.

In the present study, the RCC cohort of The Cancer Genome Atlas (TCGA) database and the ESTIMATE algorithm were both used to analyze the association of immune and stromal scores with clinicopathological features and a set of tumor microenvironment-related genes that could predict the prognosis of patients with RCC were identified. The results of the present study may provide effective genetic predictors for the diagnosis, prognosis and treatment of renal cell carcinoma.

Materials and methods

TCGA RCC data

Gene expression data for 885 patients with RCC were obtained from TCGA database (cancer.gov/tcga). In the data, 289 (32.7%) patients were female and 596 (67.3%) patients were male, with an age range of 34–90 years. The following are the selection criteria of TCGA IDs on the database: i) Disease type was adenocarcinomas; ii) primary site was the kidney; iii) program name was TCGA; iv) Workflow Type was HTSeq-FPKM; v) data category was transcriptome profiling and vi) data type was Gene Expression Quantification. RNA expression profiles of kidney adenomas and adenocarcinomas were analyzed using the Affymetrix Human Genome U133 Plus version 2.0 array. Clinical data such as sex, age, histological grading, Tumor-Node-Metastasis (TNM) stage, clinical stage, survival time and outcomes were also obtained from TCGA database.

ESTIMATE algorithm

The ESTIMATE algorithm can infer the infiltration of immune and stromal cells in tumor tissues by analyzing the transcriptional profile of cancer samples (16). Gene expression values were graded and ranked for each sample. The statistical significance value was calculated by integrating the difference between the empirical cumulative distribution functions of the signature gene and the remaining genes. The ESTIMATE algorithm output stromal and immune scores by performing single-sample Gene Set Enrichment Analysis.

Investigating the association between ESTIMATE scores and clinical features in renal cell carcinoma

Samples were grouped according to clinical/pathological stage, and the ESTIMATE scores of each group were then analyzed to evaluate the association between ESTIMATE scores and clinical/pathological stage. The mean score was used to divide these samples into low- and high-stromal/immune score groups, and the difference in overall survival rate between the two groups was evaluated using Kaplan-Meier plotter (17) and log-rank tests.

Identification of differentially expressed genes

Samples were grouped according to stromal/immune scores. The expression of individual genes was analyzed using the Limma package in R (18). All R language operations are performed in R Studio (19). A log2| fold-change (FC)| >2.0 and P<0.05 were considered to indicate a significant difference when screening for differentially expressed genes (DEGs). The Pheatmap package (CRAN.R-project.org/package=pheatmap) was used to generate heatmaps.

Enrichment analysis of differentially expressed genes

The Database for Annotation, Visualization and Integrated Discovery (DAVID v.6.8) database (david.ncifcrf.gov/) was used to identify Gene Ontology (GO) categories, including biological processes (BPs), cellular components (CCs) and molecular functions (MFs). Pathway enrichment analysis was also performed using the DAVID database with reference to the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways (kegg.jp/).

Construction of protein-protein interaction (PPI) networks

PPI networks provide a valuable framework for an improved understanding of the interaction of proteins (20). The PPI networks were retrieved from the STRING database (21) and reconstructed using Cytoscape software v.3.7.1 (22). Nodes with a high degree of connectivity in the network were considered as regulatory hubs. The node size was determined to be proportional to the degree of interaction. The genes with a degree of connectivity ≥1 were reserved in the PPI network. Genes with a degree of connectivity >31.56 were considered hub genes.

Association between the expression of DEGs and overall survival rate

The log-rank method was used to evaluate the prognostic value of each gene. Genes that were significantly associated with overall survival rate were identified as prognostic genes. Subsequently, a LASSO Cox regression model was used to construct a prognostic classifier by selecting genes from these prognostic DEGs. The risk score based on the expression levels for each patient was calculated using the constructed Cox regression model. According to the optimum cut-off score (risk score=0.472), the patients were divided into high-risk and low-risk groups. The difference in survival rates between the two groups was then assessed using the Kaplan-Meier method.

Building a clinical predictive model based on the prognostic genes for RCC

A LASSO Cox regression model was used to select the most useful prognostic genes. A multiple-gene-based classifier was constructed to predict the survival rate of patients with RCC. Patients were randomly divided into training set and testing set (7:3). Cox regression analysis was performed by using the ‘glmnet’ package. The area under the curve (AUC) of the time-dependent receiver operating characteristic (ROC) curve was used to measure the predictive accuracy of the multi-gene based classifier. Univariate and multivariate survival analyses were performed by using the Cox regression model and a nomogram plot was generated according to the Cox regression coefficients.

Statistical analysis

The differences in ESTIMATE scores between different clinical/pathological staging groups were analyzed using the Kruskal-Wallis test. The differences in the levels of gene expression between the high-and low-score groups were compared using an unpaired t-test. Kaplan-Meier analysis and log-rank tests were performed in order to compare each prognostic gene signature. Univariate and multivariate survival analyses were performed with the Cox regression model. A LASSO Cox regression analysis was used to built the best classifier according to the regression coefficients and P-values. P<0.05 was considered to indicate a statistically significant difference.

Results

ESTIMATE scores were associated with clinicopathological features in RCC

The gene expression data and clinicopathological data of 885 patients with kidney adenocarcinomas were downloaded from TCGA database. Stromal scores were distributed between −1,897.14 and 1,967.19 and immune scores ranged from 1,673.78 to 3,459.07 (data not shown). As presented in Fig. 1A-D, apart from the M stage, the immune scores were significantly associated with T, N and clinical stage (P<0.001; P<0.001; P<0.001, respectively). Similarly, the stromal scores significantly increased with increasing T, N, M and clinical stage (P=0.011; P<0.001; P<0.001; P=0.003, respectively).

Immune/stromal scores were significantly associated with overall survival rate

Patients with RCC were divided into high- and low-groups based on their scores. As presented in Fig. 1E, the overall survival rate of the low-stromal score group was more favorable compared with the high-score group (P<0.001). Patients with high immune scores should theoretically have an improved survival rate, but the statistical results revealed that the survival rate of the high-immune score group was lower compared with the low immune score group (P=0.003).

Differentially expressed genes based on immune/stromal scores

The heatmaps in Fig. 2 present the genes with significant differential expression levels between the high- and low-immune/stromal score groups. A total of 141 genes were upregulated and 277 genes were downregulated in the high-immune score group compared with the low score group (FC >2.0; P<0.05). Similarly, 327 genes were upregulated and 410 genes were downregulated in the high stromal score group compared with the low score group (FC >2.0; P<0.05). In addition, the Venn diagram demonstrated that there were 272 common DEGs shared between the high-immune score group and low-stromal score groups.

Prognostic value of differentially expressed genes in overall survival rate prediction

As presented in Table SI, among the 738 DEGs based on stromal scores, 406 DEGs were significantly associated with overall survival rate (all P<0.05). Among the 419 DEGs based on immune scores, a total of 252 DEGs were significantly associated with overall survival rate (P<0.05). Among the 272 common DEGs, a total of 137 DEGs were significantly associated with overall survival rate (P<0.05).

Functional enrichment analysis of differentially expressed genes

GO analysis of the immune scores demonstrated a strong association with the ‘immune response’ (Fig. 3A). The BP terms were primarily enriched in ‘T cell activation’, ‘response to chemokine’, ‘cellular response to chemokine’, etc. The CC terms primarily included ‘external side of plasma membrane’, ‘T cell receptor complex’, ‘side of membrane’, etc. Top MF terms included ‘chemokine receptor binding’ ‘G protein-coupled chemoattractant receptor activity’, etc. The KEGG pathway analysis revealed that a number of these pathways were associated with the immune response (Fig. 3B), including the ‘T cell receptor signaling pathway’, ‘Th1/Th2 cell differentiation’, ‘cytokine-cytokine receptor interactions’ and the ‘chemokine signaling pathway.’

Similarly, the BP terms based on stromal scores were primarily enriched in ‘extracellular structure organization’ and ‘regulation of cell migration and proliferation’ (Fig. 3C). The CC terms primarily included ‘second-messenger-mediated signaling’, ‘extracellular structure organization’, ‘extracellular matrix organization’, etc. Top MF terms included ‘extracellular matrix’, ‘collagen-containing extracellular matrix’, ‘apical part of cell’, etc. The KEGG pathway analysis identified pathways associated with ‘PI3K-Akt signaling pathway’, ‘calcium signaling’, ‘protein digestion and absorption’, ‘Rap1 signaling pathway’ and ‘cAMP signaling’ (Fig. 3D).

PPI networks and hub genes

The hub nodes in the regulation network were identified using Cytoscape software. Fig. 4A presents the PPI network of DEGs based on immune scores and displays the top 30 DEGs in the network. The top 10 noteworthy nodes were considered hub genes, including PTPRC, CCR5, CTLA4, CD2, CXCR3, CXCL9, CCL5, CCL4, CXCL10 and PRF1. Similarly, Fig. 4B presents the PPI network of DEGs based on stromal scores and the top 30 DEGs in the network. The top 10 hub genes were ADCY4, IL6, GCGR, TAC1, AGTR1, DRD2, NTS, CXCL10, GRM5 and GAL. The results of the log-rank test demonstrated that all the top 10 hub genes based on immune scores and all of the top five hub genes based on stromal scores were significantly associated with overall survival rate (Table I).

Table I.

List of hub genes that are significantly associated with overall survival rate of patients with renal cell carcinoma.

Table I.

List of hub genes that are significantly associated with overall survival rate of patients with renal cell carcinoma.

A, immune scores

Hub genesP-value
CTLA4<0.001
CCL5<0.001
CXCL9<0.001
CXCR3<0.001
CD2<0.001
PRF1<0.001
CCL4<0.001
CXCL10<0.001
CCR50.003
PTPRC0.026

B, stromal scores

Hub genesP-value

IL6<0.001
CXCL10<0.001
NTS0.014
GRM50.015
ADCY40.026

[i] Hub gene, gene whose degree of interaction was >31.56.

Building a prognostic classifier using the LASSO Cox regression model

A LASSO Cox regression model was used to construct a prognostic classifier by selecting 6 genes from the 137 common prognostic DEGs, including IL21R, ATP6V1C2, GBP1, P2RY10, GBP4 and TNNC2 (Fig. 5A). The risk score based on the expression levels of the 6 genes for each patient was calculated using the constructed Cox regression model. According to the optimum cut-off score (risk score =0.472), the patients were divided into high-risk and low-risk groups (AUC=0.776; Fig. 5B). The 6 gene based classifier was a powerful prognostic factor in the training set [hazard ratio (HR), 2.8; confidence interval (CI), 2.3–3.4; P<0.001; Fig. 5C] and in the testing set (HR, 2.3; CI, 1.6–3.4; P<0.001; Fig. 5D). The survival rate of the low-risk score group was more favorable compared with that of the high-risk score group. These results suggested that this classifier effectively predicted the prognosis of patients with RCC.

Clinical predictive model based on prognostic genes in RCC

As presented in Fig. 6A, the 6 gene based classifier demonstrated a similar prognostic accuracy to other clinical prognostic factors. The AUC values of gene based classifier, age, clinical stage and TNM stage were 0.776, 0.566, 0.822 and 0.802, respectively. The AUC of the combined classifier and clinical prognostic factors was 0.899, thus, this classifier had value in improving the accuracy of prognosis prediction of patients with RCC. To provide clinicians with a quantitative tool to predict prognosis of patients with RCC, a nomogram was constructed that combined the 6 gene based classifier and clinicopathological prognostic factors (Fig. 6B).

Discussion

RCC is one of the most common primary renal malignancies (1). Personalized data from RCC specimens can provide doctors with more accurate information to select the appropriate treatment for patients (5). The interaction between tumor cells and the tumor microenvironment substantially affects the evolution of tumors, which subsequently affects tumor recurrence, drug resistance and overall survival rate of patients (23). There is an association between the components in the tumor microenvironment and tumor outcomes (24). Different tumor microenvironmental compositions lead to differences in sensitivity to different targeted drug therapies (25). Therefore, analyzing the components in the tumor microenvironment can provide guidance for selecting the best clinical treatment options for the patient. At present, gene expression profiling has been increasingly incorporated into clinical diagnostic criteria (6). Molecular biomarkers should be considered in addition to standard clinicopathologic criteria to guide clinical treatment options.

With the rapid development of high-throughput sequencing, tumor databases with large samples, such as TCGA, provide a wealth of information to help find ways to solve difficult clinical problems, such as improving diagnostic rates and choosing targeted drugs (26). The present study aimed to identify tumor microenvironment-associated genes that were associated with the overall survival rate of patients with RCC. In the tumor microenvironment, immune and stromal cells are two major types of non-tumor components that are valuable for the diagnosis and prognosis of tumors (27). The ESTIMATE algorithm is a useful tool to extract the genetic information of immune and stromal cells from a complex tumor microenvironment in order to identify the degree of infiltration of immune and stromal cells (28). Based on ESTIMATE scores, the present study aimed to evaluate the association between gene expression signatures of immune/stromal infiltration and RCC outcomes. In particular, a set of DEGs involved in the immune response and related to the extracellular matrix were identified. GO term analysis demonstrated that the majority of these DEGs were involved in the organization and regulation of the tumor microenvironment. It has been reported that the function of immune cells and extracellular matrix molecules are involved in the establishment of the tumor microenvironment of RCC (29).

The immune components in the tumor microenvironment are complex (30). Although immune cell infiltration has been associated with response to immune checkpoint blockade in metastatic kidney cancer (31), it is difficult to predict prognosis of patients with cancer using one or several indicators. In the present study, by analyzing the clinical data, gene expression profiles and immune scores of 885 patients with RCC, it was demonstrated that the average immune scores significantly increased with increasing T, N stage and clinical stage, but were not associated with M stage. In addition, notable differences in overall survival rate based on immune gene expression signatures were observed. The presence of immune cells serves a role in killing tumor cells and inhibiting tumor growth, therefore, cases with high immune scores should theoretically have an improved survival. However, the present study demonstrated that patients with high-immune scores had less favorable survival rate when compared with those with low-immune scores. This result warrants further in-depth investigation, and such a study may identify a novel direction for immunotherapy in kidney cancer.

Non-immune cell components in the tumor microenvironment also affect the therapeutic response (32). In kidney cancer, the expression levels of transforming growth factor β signaling in fibroblasts surrounding the tumor are associated with immunologically-excluded phenotypes and a lack of response to immune checkpoint inhibition (33). Fibroblasts and other matrix components may also contribute to direct rejection or inactivation of chemotherapy (34). Based on the ESTIMATE algorithm, the present study reported that stromal scores are associated with TNM stage and clinical stage. The average stromal scores were significantly increased with increasing T stage, N, M stage and clinical stage. In addition, the overall survival rate of the low stromal score group was more favorable compared with that of the high score group. Stromal cells provide a suitable environment for tumor growth and progression (35). Analyzing the expression profiles of genes and identifying the underlying molecular mechanisms of gene expression levels affecting the tumor matrix, promoting tumorigenesis and development may provide novel insight into the clinical diagnosis and treatment of kidney cancer.

The aim of precision medicine is to implement personalized treatment based on the molecular characteristics of the patient (36). The present study aimed to identify a set of tumor microenvironment-associated genes that could predict the outcomes of patients with RCC. By comparing the gene expression levels low-score vs. high-score groups, a series of DEGs were identified. Kaplan-Meier analysis demonstrated that the 406 DEGs based on stromal scores and the 252 DEGs based on immune scores were significantly associated with overall survival rate in patients with RCC. The DEGs based on immune scores were primarily enriched in ‘immune response’ and the DEGs based on stromal scores were mainly enriched in ‘extracellular structure organization’ and the ‘regulation of cell migration and proliferation’. Notably, the present study identified 10 hub genes based on immune scores and 5 hub genes based on stromal scores that were significantly correlated with overall survival rate of patients with RCC. These novel prognostic genes could be used as potential biomarkers for the diagnosis and selection of clinical treatment options for patients with RCC.

In order to investigate the prognostic value of these DEGS, a LASSO Cox regression model was used to construct a 6 gene based classifier, which was demonstrated to be an effective prognostic factor. The results of the statistical analysis demonstrated that the survival rate in patients with low-risk scores was higher compared with patients with high-risk scores. These results indicate that the 6 gene based classifier had high accuracy in predicting the prognosis of patients with RCC. Furthermore, combining this classifier with other clinical variables could lead to more accurate predictions of patient survival. A nomogram combining this classifier and clinical risk factors may aid clinicians in the prediction of prognosis of patients with RCC.

In summary, the immune/stromal gene signatures of RCC are associated with clinicopathological factors and overall survival rate. The present study identified a set of tumor microenvironment-associated genes that were associated with the prognosis of patients with RCC. The classifier based on these tumor microenvironment-associated genes had prognostic value in predicting the prognosis of patients with RCC. Further study of these genes may provide a comprehensive understanding of the association between the tumor microenvironment and the prognosis of RCC. Although validation in prospective trials is necessary, the results of the present study suggest that transcriptional analysis may be valuable to predict the prognosis of patients with RCC.

Supplementary Material

Supporting Data

Acknowledgements

The authors would like to thank Dr. Chen (Sun Yat-sen University, China) for providing R language software.

Funding

The present study was supported by The National Natural Science Foundation of China (grant no. 81702516).

Availability of data and materials

The results published in the present study are based upon data generated by the TCGA Research Network (http://cancergenome.nih.gov/).

Authors contributions

SSC contributed to conception, data analysis, and final approval of the version to be published. SW contributed to the data analysis. XGZ and XLC made substantial contributions to data analysis and revising the manuscript critically for important intellectual content. XJS and SSC were involved in drafting the manuscript and contributed to the data analysis. 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:

RCC

renal cell carcinoma

DEGs

differentially expressed genes

GO

Gene Ontology

BP

biological processes

MF

molecular functions

CC

cellular components

KEGG

Kyoto Encyclopedia of Genes and Genomes

PPI

protein-protein interaction

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2019. CA Cancer J Clin. 69:7–34. 2019. View Article : Google Scholar : PubMed/NCBI

2 

Zhang Q, Shi J, Yuan F, Wang H, Fu W, Pan J, Huang Y, Yu J, Yang J and Chen Z: Higher expression of XPF is a critical factor in intrinsic chemotherapy resistance of human renal cell carcinoma. Int J Cancer. 139:2827–2837. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Dos Santos M, Brachet PE, Chevreau C and Joly F: Impact of targeted therapies in metastatic renal cell carcinoma on patient-reported outcomes: Methodology of clinical trials and clinical benefit. Cancer Treat Rev. 53:53–60. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Chen L, Yuan L, Qian K, Qian G, Zhu Y, Wu CL, Dan HC, Xiao Y and Wang X: Identification of biomarkers associated with pathological stage and prognosis of clear cell renal cell carcinoma by co-expression network analysis. Front Physiol. 9:3992018. View Article : Google Scholar : PubMed/NCBI

5 

Goto-Yamaguchi L, Yamamoto-Ibusuki M, Yamamoto Y, Fujiki Y, Tomiguchi M, Sueta A, Takeshita T and Iwase H: Therapeutic predictors of neoadjuvant endocrine therapy response in estrogen receptor-positive breast cancer with reference to optimal gene expression profiling. Breast Cancer Res Treat. 172:353–362. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Tse LA, Dai J, Chen M, Liu Y, Zhang H, Wong TW, Leung CC, Kromhout H, Meijer E, Liu S, et al: Prediction models and risk assessment for silicosis using a retrospective cohort study among workers exposed to silica in China. Sci Rep. 5:110592015. View Article : Google Scholar : PubMed/NCBI

7 

Gatto F, Blum KA, Hosseini SS, Ghanaat M, Kashan M, Maccari F, Galeotti F, Hsieh JJ, Volpi N, Hakimi AA, et al: Plasma glycosaminoglycans as diagnostic and prognostic biomarkers in surgically treated eenal cell carcinoma. Eur Urol Oncol. 1:364–377. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Wang SC, Wang ST, Liu HT, Wang XY, Wu SC, Chen LC and Liu YW: Trichostatin A induces bladder cancer cell death via intrinsic apoptosis at the early phase and Sp1-survivin downregulation at the late phase of treatment. Oncol Rep. 38:1587–1596. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Sweis RF, Spranger S, Bao R, Paner GP, Stadler WM, Steinberg G and Gajewski TF: Molecular drivers of the non-T-cell-inflamed tumor microenvironment in urothelial bladder cancer. Cancer Immunol Res. 4:563–568. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Chen F, Zhang Y, Bossé D, Lalani AA, Hakimi AA, Hsieh JJ, Choueiri TK, Gibbons DL, Ittmann M and Creighton CJ: Pan-urologic cancer genomic subtypes that transcend tissue of origin. Nat Commun. 8:1992017. View Article : Google Scholar : PubMed/NCBI

11 

Maniecki MB, Etzerodt A, Ulhøi BP, Steiniche T, Borre M, Dyrskjøt L, Orntoft TF, Moestrup SK and Møller HJ: Tumor-promoting macrophages induce the expression of the macrophage-specific receptor CD163 in malignant cells. Int J Cancer. 131:2320–2331. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Tsai S, McOlash L, Palen K, Johnson B, Duris C, Yang Q, Dwinell MB, Hunt B, Evans DB, Gershan J, et al: Development of primary human pancreatic cancer organoids, matched stromal and immune cells and 3D tumor microenvironment models. BMC Cancer. 18:3352018. View Article : Google Scholar : PubMed/NCBI

13 

Garcia-Gomez A, Rodríguez-Ubreva J and Ballestar E: Epigenetic interplay between immune, stromal and cancer cells in the tumor microenvironment. Clin Immunol. 196:64–71. 2018. View Article : Google Scholar : PubMed/NCBI

14 

Jia D, Li S, Li D, Xue H, Yang D and Liu Y: Mining TCGA database for genes of prognostic value in glioblastoma microenvironment. Aging (Albany NY). 10:592–605. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Gan TQ, Chen WJ, Qin H, Huang SN, Yang LH, Fang YY, Pan LJ, Li ZY and Chen G: Clinical value and prospective pathway signaling of microRNA-375 in lung adenocarcinoma: A study based on the Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO) and bioinformatics analysis. Med Sci Monit. 23:2453–2464. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Yoshihara K, Shahmoradgoli M, Martínez E, Vegesna R, Kim H, Torres-Garcia W, Treviño V, Shen H, Laird PW, Levine DA, et al: Inferring tumour purity and stromal and immune cell admixture from expression data. Nat Commun. 4:26122013. View Article : Google Scholar : PubMed/NCBI

17 

Nagy Á, Lánczky A, Menyhárt O and Győrffy B: Validation of miRNA prognostic power in hepatocellular carcinoma using expression data of independent datasets. Sci Rep. 8:92272018. View Article : Google Scholar : PubMed/NCBI

18 

R Core Team, . 2012.R: A language and environment for statistical computing. R Foundation for Statistical Computing. (Vienna, Austria). Available online at https://www.R-project.org/.

19 

RStudio Team, . RStudio: Integrated Development for R. RStudio, Inc.; Boston, MA: 2015, http://www.rstudio.com/

20 

Haque M, Sarmah R and Bhattacharyya DK: A common neighbor based technique to detect protein complexes in PPI networks. J Genet Eng Biotechnol. 16:227–238. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, Simonovic M, Doncheva NT, Morris JH, Bork P, et al: STRING v11: Protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 47(D1): D607–D613. 2019. View Article : Google Scholar : PubMed/NCBI

22 

Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B and Ideker T: Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res. 13:2498–2504. 2003. View Article : Google Scholar : PubMed/NCBI

23 

Patel H, Nilendu P, Jahagirdar D, Pal JK and Sharma NK: Modulating secreted components of tumor microenvironment: A masterstroke in tumor therapeutics. Cancer Biol Ther. 19:3–12. 2018. View Article : Google Scholar : PubMed/NCBI

24 

Chevrier S, Levine JH, Zanotelli VRT, Silina K, Schulz D, Bacac M, Ries CH, Ailles L, Jewett MAS, Moch H, et al: An Immune Atlas of Clear Cell Renal Cell Carcinoma. Cell. 169:736–749.e18. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Herting CJ, Chen Z, Pitter KL, Szulzewsky F, Kaffes I, Kaluzova M, Park JC, Cimino PJ, Brennan C, Wang B, et al: Genetic driver mutations define the expression signature and microenvironmental composition of high-grade gliomas. Glia. 65:1914–1926. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Deng M, Brägelmann J, Schultze JL and Perner S: Web-TCGA: An online platform for integrated analysis of molecular cancer data sets. BMC Bioinformatics. 17:722016. View Article : Google Scholar : PubMed/NCBI

27 

Andreeva E, Bobyleva P, Gornostaeva A and Buravkova L: Interaction of multipotent mesenchymal stromal and immune cells: Bidirectional effects. Cytotherapy. 19:1152–1166. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Schelker M, Feau S, Du J, Ranu N, Klipp E, MacBeath G, Schoeberl B and Raue A: Estimation of immune cell content in tumour tissue using single-cell RNA-seq data. Nat Commun. 8:20322017. View Article : Google Scholar : PubMed/NCBI

29 

Ghatalia P, Gordetsky J, Kuo F, Dulaimi E, Cai KQ, Devarajan K, Bae S, Naik G, Chan TA, Uzzo R, et al: Prognostic impact of immune gene expression signature and tumor infiltrating immune cells in localized clear cell renal cell carcinoma. J Immunother Cancer. 7:1392019. View Article : Google Scholar : PubMed/NCBI

30 

Dai J, Lu Y, Roca H, Keller JM, Zhang J, McCauley LK and Keller ET: Immune mediators in the tumor microenvironment of prostate cancer. Chin J Cancer. 36:292017. View Article : Google Scholar : PubMed/NCBI

31 

Kamal Y, Cheng C, Frost HR and Amos CI: Predictors of disease aggressiveness influence outcome from immunotherapy treatment in renal clear cell carcinoma. OncoImmunology. 8:e15001062018. View Article : Google Scholar : PubMed/NCBI

32 

Faurobert E, Bouin AP and Albiges-Rizo C: Microenvironment, tumor cell plasticity, and cancer. Curr Opin Oncol. 27:64–70. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Wang L, Wen W, Yuan J, Helfand B, Li Y, Shi C, Tian F, Zheng J, Wang F, Chen L, et al: Immunotherapy for human renal cell carcinoma by adoptive transfer of autologous transforming growth factor beta-insensitive CD8+ T cells. Clin Cancer Res. 16:164–173. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Li M, Li M, Yin T, Shi H, Wen Y, Zhang B, Chen M, Xu G, Ren K and Wei Y: Targeting of cancer-associated fibroblasts enhances the efficacy of cancer chemotherapy by regulating the tumor microenvironment. Mol Med Rep. 13:2476–2484. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Raguraman R, Parameswaran S, Kanwar JR, Khetan V, Rishi P, Kanwar RK and Krishnakumar S: Evidence of tumour microenvironment and stromal cellular components in retinoblastoma. Ocul Oncol Pathol. 5:85–93. 2019. View Article : Google Scholar : PubMed/NCBI

36 

Zhang J, Späth SS, Marjani SL, Zhang W and Pan X: Characterization of cancer genomic heterogeneity by next-generation sequencing advances precision medicine in cancer treatment. Precis Clin Med. 1:29–48. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2020
Volume 20 Issue 1

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
Wang S, Zheng X, Chen X, Shi X and Chen S: Prognostic and predictive value of immune/stromal-related gene biomarkers in renal cell carcinoma. Oncol Lett 20: 308-316, 2020
APA
Wang, S., Zheng, X., Chen, X., Shi, X., & Chen, S. (2020). Prognostic and predictive value of immune/stromal-related gene biomarkers in renal cell carcinoma. Oncology Letters, 20, 308-316. https://doi.org/10.3892/ol.2020.11574
MLA
Wang, S., Zheng, X., Chen, X., Shi, X., Chen, S."Prognostic and predictive value of immune/stromal-related gene biomarkers in renal cell carcinoma". Oncology Letters 20.1 (2020): 308-316.
Chicago
Wang, S., Zheng, X., Chen, X., Shi, X., Chen, S."Prognostic and predictive value of immune/stromal-related gene biomarkers in renal cell carcinoma". Oncology Letters 20, no. 1 (2020): 308-316. https://doi.org/10.3892/ol.2020.11574